Language selection

Search

Patent 3022327 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3022327
(54) English Title: SYNTHESIS OF INDAZOLES
(54) French Title: SYNTHESE D'INDAZOLES
Status: Allowed
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07D 401/12 (2006.01)
  • A61K 31/4439 (2006.01)
  • A61P 9/00 (2006.01)
  • A61P 27/00 (2006.01)
  • A61P 35/00 (2006.01)
  • A61P 37/00 (2006.01)
(72) Inventors :
  • THALER, TOBIAS (Germany)
  • PLATZEK, JOHANNES (Germany)
  • GUIMOND, NICOLAS (Germany)
(73) Owners :
  • BAYER PHARMA AKTIENGESELLSCHAFT (Germany)
(71) Applicants :
  • BAYER PHARMA AKTIENGESELLSCHAFT (Germany)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2017-04-25
(87) Open to Public Inspection: 2017-11-02
Examination requested: 2022-04-22
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/EP2017/059748
(87) International Publication Number: WO2017/186693
(85) National Entry: 2018-10-26

(30) Application Priority Data:
Application No. Country/Territory Date
16167650.7 European Patent Office (EPO) 2016-04-29
16167649.9 European Patent Office (EPO) 2016-04-29

Abstracts

English Abstract

The present invention relates to a novel method of preparing a 2-substituted indazole of formula (I) to novel intermediate compounds, and to the use of intermediate compounds for the preparation of said 2-substituted indazole.


French Abstract

La présente invention concerne un nouveau procédé de préparation d'indazole 2-substitué de formule (I), des nouveaux composés intermédiaires, et l'utilisation de ces composés intermédiaires pour la préparation dudit indazole 2-substitué.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
1. A method of preparing a compound of formula (l):
Image
comprising the following step (A):
wherein a compound of formula (lla):
Image
is allowed to react with a compound of formula (Vl):
Image
optionally in the presence of an organic base, particularly a weak base, such
as a tertiary amine,
such as N,N-diisopropylethylamine for example,
optionally in an aromatic hydrocarbon solvent, such as toluene, xylene and
mesitylene for
example,
thereby providing said compound of formula (l).


2. The method according to claim 1, wherein said aromatic hydrocarbon solvent
is toluene.
3. The method according to claim 1 or 2, wherein said organic base is N,N-
diisopropylethylamine.
4. The method according to claim 1, 2 or 3 wherein said compound of formula
(lla):
Image
is prepared by the following step (B):
wherein a compound of formula (VIla):
Image
is allowed to react with a reductive methylating agent, such as a
methylmetallic agent, such as a
methylmagnesium halide, such as methylmagnesium chloride for example,
optionally in the presence of an alkali metal halide, such as lithium chloride
for example,
thereby providing said compound of formula (lla).
5. The method according to any one of claims 1 to 4, wherein said compound of
formula (VIla):

71

Image
is prepared by the following step (C):
wherein a compound of formula (XII):
Image
is allowed to react with a compound of formula (IX):
Image
optionally in the presence of an organic base, particularly a weak organic
base, such as a tertiary
amine, such as N,N-diisopropylethylamine for example,
optionally in the presence of a coupling agent, such as 2,4,6-tripropyl-
1,3,5,2,4,6-
trioxatriphosphinane 2,4,6-trioxide (T3P) for example,
thereby providing said compound of formula (VIla).

72

6. The method according to any one of claims 1 to 5, wherein said compound of
formula (I) is
prepared via the following steps shown in reaction scheme IA, infra:
Image
7. The method according to any one of claims 1 to 6, wherein said compound of
formula (I) is
prepared via the following steps shown in reaction scheme I, infra:
Image
8. The method according to any one of claims 1 to 7, wherein said compound of
formula (I) is purified
by crystallization, particularly from a solvent or a mixture of solvents such
as a mixture of acetone
and toluene, optionally in the presence of activated charcoal, optionally
followed by a further
crystallization from a solvent such as ethanol for example.
9. The method according to claim 8, wherein said compound of formula (I) is in
the form of crystalline
needles (A).
10. Use of a compound selected from:
73

Image
for preparing a compound of formula (l):
Image
by the method according to any one of claims 1 to 9.
11. Use of a compound of structure:
Image
for preparing a compound of formula (l):
74

Image

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03022327 2018-10-26
WO 2017/186693
APIPCT/EP2017/059748 FC
SYNTHESIS of INDAZOLES
The present invention relates to a novel method of preparing a 2-substituted
indazole with the
following structure:
F3c
N ( OH
HO
(I)
to a novel crystalline needle form of said 2-substituted indazole, to novel
intermediate compounds,
and to the use of intermediate compounds for the preparation of said 2-
substituted indazole.
The present invention relates to the preparation of substituted indazole of
formula (I) which inhibits
interleukin-1 receptor-associated kinase 4 (IRAK4).
Human IRAK4 (interleukin-1 receptor-associated kinase 4) plays a key role in
the activation of the
immune system. Therefore, this kinase is an important therapeutic target
molecule for the
development of inflammation-inhibiting substances. IRAK4 is expressed by a
multitude of cells and
mediates the signal transduction of Toll-like receptors (TLR), except TLR3,
and receptors of the
interleukin (IL)-113 family consisting of the IL-1R (receptor), IL-18R, IL-33R
and IL-36R (Janeway and
Medzhitov, Annu. Rev. Immunol., 2002; Dinarello, Annu. Rev. Immunol., 2009;
Flannery and Bowie,
Biochemical Pharmacology, 2010).
Neither IRAK4 knockout mice nor human cells from patients lacking IRAK4 react
to stimulation by
TLRs (except TLR3) and the IL-13 family (Suzuki, Suzuki, et al., Nature, 2002;
Davidson, Currie, et al.,
The Journal of Immunology, 2006; Ku, von Bernuth, et al., JEM, 2007; Kim,
Staschke, et al., JEM,
2007).
The binding of the TLR ligands or the ligands of the IL-13 family to the
respective receptor leads to
recruitment and binding of MyD88 [Myeloid differentiation primary response
gene (88)] to the
receptor. As a result, MyD88 interacts with IRAK4, resulting in the formation
of an active complex
which interacts with and activates the kinases IRAK1 or IRAK2 (Kollewe,
Mackensen, et al., Journal of
Biological Chemistry, 2004; Precious et al., J. Biol. Chem., 2009). As a
result of this, the NF (nuclear
factor)-kB signalling pathway and the MAPK (mitogen-activated protein kinase)
signal pathway is
1

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
activated (Wang, Deng, et al., Nature, 2001). The activation both of the NF-kB
signalling pathway and
of the MAPK signalling pathway leads to processes associated with different
immune processes. For
example, there is increased expression of various inflammatory signal
molecules and enzymes such
as cytokines, chemokines and COX-2 (cyclooxygenase-2), and increased mRNA
stability of
inflammation-associated genes, for example COX-2, IL-6, IL-8 (Holtmann,
Enninga, et al., Journal of
Biological Chemistry, 2001; Datta, Novotny, et al., The Journal of Immunology,
2004). Furthermore,
these processes may be associated with the proliferation and differentiation
of particular cell types,
for example monocytes, macrophages, dendritic cells, T cells and B cells (Wan,
Chi, et al., Nat
Immunol, 2006; McGettrick and J. O'Neill, British Journal of Haematology,
2007).
The central role of IRAK4 in the pathology of various inflammatory disorders
had already been shown
by direct comparison of wild-type (WT) mice with genetically modified animals
having a kinase-
inactivated form of IRAK4 (IRAK4 KDKI). IRAK4 KDKI animals have an improved
clinical picture in the
animal model of multiple sclerosis, atherosclerosis, myocardial infarction and
Alzheimer's disease
(Rekhter, Staschke, et al., Biochemical and Biophysical Research
Communication, 2008; Maekawa,
Mizue, et al., Circulation, 2009; Staschke, Dong, et al., The Journal of
Immunology, 2009; Kim,
Febbraio, et al., The Journal of Immunology, 2011; Cameron, Tse, et al., The
Journal of Neuroscience,
2012). Furthermore, it was found that deletion of IRAK4 in the animal model
protects against virus-
induced myocarditis an improved anti-viral reaction with simultaneously
reduced systemic
inflammation (Valaperti, Nishii, et al., Circulation, 2013). It has also been
shown that the expression
of IRAK4 correlates with the degree of Vogt-Koyanagi-Harada syndrome (Sun,
Yang, et al., PLoS ONE,
2014).
As well as the essential role of IRAK4 in congenital immunity, there are also
hints that IRAK4
influences the differentiation of what are called the Th17 T cells, components
of adaptive immunity.
In the absence of IRAK4 kinase activity, fewer IL-17-producing T cells (Th17 T
cells) are generated
compared to WT mice. The inhibition of IRAK4 is therefore suitable for
prophylaxis and/or treatment
of atherosclerosis, type 1 diabetes, rheumatoid arthritis, spondyloarthritis,
lupus erythematosus,
psoriasis, vitiligo, chronic inflammatory bowel disease and viral disorders,
for example HIV (human
immunodeficiency virus), hepatitis virus (Staschke, et al., The Journal of
Immunology, 2009;
Zambrano-Zaragoza, et al., International Journal of Inflammation, 2014).
Owing to the central role of IRAK4 in the MyD88-mediated signal cascade of
TLRs (except TLR3) and
the IL-1 receptor family, the inhibition of IRAK4 can be utilized for the
prophylaxis and/or treatment
of disorders mediated by the receptors mentioned. TLRs and also components of
the IL-1 receptor
2

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
family are involved in the pathogenesis of rheumatoid arthritis, metabolic
syndrome, diabetes,
osteoarthritis, Sjogren syndrome and sepsis (Scanzello, Plaas, et al. Curr
Opin Rheumatol, 2008;
Roger, Froidevaux, et al, PNAS, 2009; Gambuzza, Licata, et al., Journal of
Neuroimmunology, 2011;
Fresno, Archives Of Physiology And Biochemistry, 2011; Volin and Koch, J
Interferon Cytokine Res,
2011; Akash, Shen, et al., Journal of Pharmaceutical Sciences, 2012; Goh and
Midwood,
Rheumatology, 2012; Dasu, Ramirez, et al., Clinical Science, 2012; Ramirez and
Dasu, Curr Diabetes
Rev, 2012; Li, Wang, et al., Pharmacology & Therapeutics, 2013; Sedimbi,
Hagglof, et al., Cell Mol Life
Sci, 2013; Talabot-Aye, et al., Cytokine, 2014). Skin diseases such as
psoriasis, atopic dermatitis,
Kindler's syndrome, allergic contact dermatitis, acne inversa and acne
vulgaris are associated with
the IRAK4-mediated TLR signalling pathway (Gilliet, Conrad, et al., Archives
of Dermatology, 2004;
Niebuhr, Langnickel, et al., Allergy, 2008; Miller, Adv Dermatol, 2008;
Terhorst, Kalali, et al., Am J Clin
Dermatol, 2010; Viguier, Guigue, et al., Annals of Internal Medicine, 2010;
Cevikbas, Steinhoff, J
Invest Dermatol, 2012; Minkis, Aksentijevich, et al., Archives of Dermatology,
2012; Dispenza,
Wolpert, et al., J Invest Dermatol, 2012; Minkis, Aksentijevich, et al.,
Archives of Dermatology, 2012;
Gresnigt and van de Veerdonk, Seminars in Immunology, 2013; Selway, Kurczab,
et al., BMC
Dermatology, 2013; Sedimbi, Hagglof, et al., Cell Mol Life Sci, 2013; Wollina,
Koch, et al. Indian
Dermatol Online, 2013; Foster, Baliwag, et al., The Journal of Immunology,
2014).
Pulmonary disorders such as pulmonary fibrosis, obstructive pulmonary disease
(COPD), acute
respiratory distress syndrome (ARDS), acute lung injury (ALI), interstitial
lung disease (ILD),
sarcoidosis and pulmonary hypertension also show an association with various
TLR-mediated
signalling pathways. The pathogenesis of the pulmonary disorders may be either
infectiously
mediated or non-infectiously mediated processes (Ramirez Cruz, Maldonado
Bernal, et al., Rev Alerg
Mex, 2004; Jeyaseelan, Chu, et al., Infection and Immunity, 2005; Seki,
Tasaka, et al., Inflammation
Research, 2010; Xiang, Fan, et al., Mediators of Inflammation, 2010;
Margaritopoulos, Antoniou, et
al., Fibrogenesis & Tissue Repair, 2010; Hilberath, Carlo, et al., The FASEB
Journal, 2011; Nadigel,
Prefontaine, et al., Respiratory Research, 2011; Kovach and Standiford,
International
Immunopharmacology, 2011; Bauer, Shapiro, et al., Mol Med, 2012; Deng, Yang,
et al., PLoS One,
2013; Freeman, Martinez, et al., Respiratory Research, 2013; Dubaniewicz, A.,
Human Immunology,
.. 2013). TLRs and also IL-1R family members are also involved in the
pathogenesis of other
inflammatory disorders such as Behget's disease, gout, lupus erythematosus,
adult-onset Still's
disease and chronic inflammatory bowel diseases such as ulcerative colitis and
Crohn's disease, and
transplant rejection, and so inhibition of IRAK4 here is a suitable
therapeutic approach (Liu-Bryan,
Scott, et al., Arthritis & Rheumatism, 2005; Christensen, Shupe, et al.,
Immunity, 2006; Cario,
Inflammatory Bowel Diseases, 2010; Nickerson, Christensen, et al., The Journal
of Immunology, 2010;
3

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Rakoff-Nahoum, Hao, et al., Immunity, 2006; Heimesaat, Fischer, et al., PLoS
ONE, 2007; Kobori, Yagi,
et al., J Gastroenterol, 2010; Shi, Mucsi, et al., Immunological Reviews,
2010; Leventhal and
Schroppel, Kidney Int, 2012; Chen, Lin, et al., Arthritis Res Ther, 2013; Hao,
Liu, et al., Curr Opin
Gastroenterol, 2013; Kreisel and Goldstein, Transplant International, 2013;
Li, Wang, et al.,
Pharmacology & Therapeutics, 2013; Walsh, Carthy, et al., Cytokine & Growth
Factor Reviews, 2013;
Zhu, Jiang, et al., Autoimmunity, 2013; Yap and Lai, Nephrology, 2013).
Because of the mechanism of
action of the compound of formula (I), they are also suitable for prophylactic
and/or therapeutic use
of the TLR and IL-1R family-mediated disorders endometriosis and
atherosclerosis (Akoum, Lawson,
et al., Human Reproduction, 2007; Allhorn, Boing, et al., Reproductive Biology
and Endocrinology,
2008; Lawson, Bourcier, et al., Journal of Reproductive Immunology, 2008;
Seneviratne,
Sivagurunathan, et al., Clinica Chimica Acta, 2012; Sikora, Mielczarek-Palacz,
et al., American Journal
of Reproductive Immunology, 2012; Falck-Hansen, Kassiteridi, et al.,
International Journal of
Molecular Sciences, 2013; Khan, Kitajima, et al., Journal of Obstetrics and
Gynaecology Research,
2013; Santulli, Borghese, et al., Human Reproduction, 2013; Sedimbi, Hagglof,
et al., Cell Mol Life Sci,
2013).
In addition to the disorders already mentioned, IRAK4-mediated TLR processes
have been described
in the pathogenesis of eye disorders such as retinal ischaemia, keratitis,
allergic conjunctivitis,
keratoconjunctivitis sicca, macular degeneration and uveitis (Kaarniranta and
Salminen, J Mol Med
(Berl), 2009; Sun and Pearlman, Investigative Ophthalmology & Visual Science,
2009; Redfern and
McDermott, Experimental Eye Research, 2010; Kezic, Taylor, et al., J Leukoc
Biol, 2011; Chang,
McCluskey, et al., Clinical & Experimental Ophthalmology, 2012; Guo, Gao, et
al., Immunol Cell Biol,
2012; Lee, Hattori, et al., Investigative Ophthalmology & Visual Science,
2012; Qi, Zhao, et al.,
Investigative Ophthalmology & Visual Science, 2014).
Because of the central role of IRAK4 in TLR-mediated processes, the inhibition
of IRAK4 also enables
the treatment and/or prevention of cardiovascular and neurological disorders,
for example
myocardial reperfusion damage, myocardial infarction, hypertension (Oyama,
Blais, et al.,
Circulation, 2004; Timmers, Sluijter, et al., Circulation Research, 2008; Fang
and Hu, Med Sci Monit,
2011; Bijani, International Reviews of Immunology, 2012; Bomfim, Dos Santos,
et al., Clin Sci (Lond),
2012; Christia and Frangogiannis, European Journal of Clinical Investigation,
2013; Thompson and
Webb, Clin Sci (Lond), 2013;), and also Alzheimer's disease, stroke,
craniocerebral trauma and
Parkinson's disease (Brough, Tyrrell, et al., Trends in Pharmacological
Sciences, 2011; Carty and
Bowie, Biochemical Pharmacology, 2011; Denes, Kitazawa, Cheng, et al., The
Journal of Immunology,
2011; Lim, Kou, et al., The American Journal of Pathology, 2011; Beraud and
Maguire-Zeiss,
4

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Parkinsonism & Related Disorders, 2012; Denes, Wilkinson, et al., Disease
Models & Mechanisms,
2013; Noelker, Morel, et al., Sci. Rep., 2013; Wang, Wang, et al., Stroke,
2013).
Because of the involvement of TLR signals and IL-1 receptor family-mediated
signals via IRAK4 in the
case of pruritus and pain, for example cancer pain, post-operative pain,
inflammation-induced and
chronic pain, there may be assumed to be a therapeutic effect in the
indications mentioned through
the inhibition of IRAK4 (Wolf, Livshits, et al., Brain, Behavior, and
Immunity, 2008; Kim, Lee, et al.,
Toll-like Receptors: Roles in Infection and Neuropathology, 2009; del Rey,
Apkarian, et al., Annals of
the New York Academy of Sciences, 2012; Guerrero, Cunha, et al., European
Journal of
Pharmacology, 2012; Kwok, Hutchinson, et al., PLoS ONE, 2012; Nicotra, Loram,
et al., Experimental
Neurology, 2012; Chopra and Cooper, J Neuroimmune Pharmacol, 2013; David,
Ratnayake, et al.,
Neurobiology of Disease, 2013; Han, Zhao, et al., Neuroscience, 2013; Liu and
Ji, Pflugers Arch., 2013;
Stokes, Cheung, et al., Journal of Neuroinflammation, 2013; Zhao, Zhang, et
al., Neuroscience, 2013;
Liu, Y. Zhang, et al., Cell Research, 2014).
This also applies to some oncological disorders. Particular lymphomas, for
example ABC-DLBCL
(activated B-cell diffuse large-cell B-cell lymphoma), mantle cell lymphoma
and Waldenstrom's
disease, and also chronic lymphatic leukaemia, melanoma and liver cell
carcinoma, are characterized
by mutations in MyD88 or changes in MyD88 activity which can be treated by an
IRAK4 inhibitor
(Ngo, Young, et al., Nature, 2011; Puente, Pinyol, et al., Nature, 2011;
Srivastava, Geng, et al., Cancer
Research, 2012; Treon, Xu, et al., New England Journal of Medicine, 2012;
Choi, Kim, et al., Human
Pathology, 2013; (Liang, Chen, et al., Clinical Cancer Research, 2013). In
addition, MyD88 plays an
important role in ras-dependent tumours, and so IRAK4 inhibitors are also
suitable for treatment
thereof (Kfoury, A., K. L. Corf, et al., Journal of the National Cancer
Institute, 2013).
Inflammatory disorders such as CAPS (cryopyrin-associated periodic syndromes)
including FCAS
(familial cold autoinflammatory syndrome), MWS (Muckle-Wells syndrome), NOMID
(neonatal-onset
multisystem inflammatory disease) and CONCA (chronic infantile, neurological,
cutaneous, and
articular) syndrome; FMF (familial mediterranean fever), HIDS (hyper-IgD
syndrome), TRAPS (tumour
necrosis factor receptor 1-associated periodic syndrom), juvenile idiopathic
arthritis, adult-onset
Still's disease, Adamantiades-Behget's disease,
rheumatoid arthritis, osteoarthritis,
keratoconjunctivitis sicca and Sjogren syndrome are treated by blocking the IL-
1 signal pathway;
therefore here, too, an IRAK4 inhibitor is suitable for treatment of the
diseases mentioned
(Narayanan, Corrales, et al., Cornea, 2008; Henderson and Goldbach-Mansky,
Clinical Immunology,
2010; Dinarello, European Journal of Immunology, 2011; Gul, Tugal-Tutkun, et
al., Ann Rheum Dis,
2012; Pettersson, Annals of MedicinePetterson, 2012; Ruperto, Brunner, et al.,
New England Journal
5

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
of Medicine, 2012; Nordstrom, Knight, et al., The Journal of Rheumatology,
2012; Vijmasi, Chen, et
al., Mol Vis, 2013; Yamada, Arakaki, et al., Opinion on Therapeutic Targets,
2013). The ligand of IL-
33R, IL-33, is involved particularly in the pathogenesis of acute kidney
failure, and so the inhibition of
IRAK4 for prophylaxis and/or treatment is a suitable therapeutic approach
(Akcay, Nguyen, et al.,
Journal of the American Society of Nephrology, 2011). Components of the IL-1
receptor family are
associated with myocardial infarction, different pulmonary disorders such as
asthma, COPD,
idiopathic interstitial pneumonia, allergic rhinitis, pulmonary fibrosis and
acute respiratory distress
syndrome (ARDS), and so prophylactic and/or therapeutic action is to be
expected in the indications
mentioned through the inhibition of IRAK4 (Kang, Homer, et al., The Journal of
Immunology, 2007;
Imaoka, Hoshino, et al., European Respiratory Journal, 2008; Couillin,
Vasseur, et al., The Journal of
Immunology, 2009; Abbate, Kontos, et al., The American Journal of Cardiology,
2010; Lloyd, Current
Opinion in Immunology, 2010; Pauwels, Bracke, et al., European Respiratory
Journal, 2011; Haenuki,
Matsushita, et al., Journal of Allergy and Clinical Immunology, 2012; Yin, Li,
et al., Clinical &
Experimental Immunology, 2012; Abbate, Van Tassel!, et al., The American
Journal of Cardiology,
2013; Alexander-Brett, et al., The Journal of Clinical Investigation, 2013;
Bunting, Shadie, et al.,
BioMed Research International, 2013; Byers, Alexander-Brett, et al., The
Journal of Clinical
Investigation, 2013; Kawayama, Okamoto, et al., J Interferon Cytokine Res,
2013; Martinez-Gonzalez,
Roca, et al., American Journal of Respiratory Cell and Molecular Biology,
2013; Nakanishi, Yamaguchi,
et al., PLoS ONE, 2013; Qiu, Li, et al., Immunology, 2013; Li, Guabiraba, et
al., Journal of Allergy and
Clinical Immunology, 2014; Saluja, Ketelaar, et al., Molecular Immunology,
2014).
The prior art discloses a multitude of IRAK4 inhibitors (see, for example,
Annual Reports in Medicinal
Chemistry (2014), 49, 117 ¨ 133).
U58293923 and U520130274241 disclose IRAK4 inhibitors having a 3-substituted
indazole structure.
.. There is no description of 2-substituted indazoles.
W02013/106254 and W02011/153588 disclose 2,3-disubstituted indazole
derivatives.
W02007/091107 describes 2-substituted indazole derivatives for the treatment
of Duchenne
muscular dystrophy. The compounds disclosed do not have 6-hydroxyalkyl
substitution.
W02015/091426 describes indazoles, the alkyl group thereof substituted at
position 2 by a
carboxamide structure.
W02015/104662 disloses indazole compounds of formula (I)
6

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
(R n H
Nl 0
N RI
(I)
/
which are therapeutically useful as kinase inhibitor, particularly IRAK4
inhibitors, and
pharmaceutically acceptable salts or stereoisomers thereof that are useful in
the treatment and
prevention of diseases or disorder, in particular their use in diseases or
disorder mediated by kinase
enzyme, particularly IRAK4 enzyme.
W02016/083433, published after the priority date of the present application,
describes novel
substituted indazoles of the following formula
Ire\ _
>c
I
1,24NrC)
HN
---- N-R1
N
R2 R3 /
methods for the production thereof, use thereof alone or in combinations to
treat and/or prevent
diseases, and use thereof to produce drugs for treating and/or preventing
diseases, in particular for
treating and/or preventing endometriosis and endometriosis-associated pain and
other symptoms
associated with endometriosis such as dysmenorrhea, dyspareunia, dysuria, and
dyschezia,
lymphomas, rheumatoid arthritis, spondyloarthritides (in particular psoriatic
spondyloarthritis and
Bekhterev's disease), lupus erythematosus, multiple sclerosis, macular
degeneration, COPD, gout,
fatty liver diseases, insulin resistance, tumor diseases, and psoriasis.
The novel IRAK4 inhibitor shall be especially suitable for treatment and for
prevention of proliferative
and inflammatory disorders characterized by an overreacting immune system.
Particular mention
should be made here of inflammatory skin disorders, cardiovascular disorders,
lung disorders, eye
disorders, autoimmune disorders, gynaecological disorders, especially
endometriosis, and cancer.
A process was to be disclosed that would allow the production of indazole (I)
on technical scale with
special focus on the following requirements:
= Scale-up/scalability of the manufacturing process
= High regioselectivity in the N2-alkylation reaction
7

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
= Process safety
= Speed of production
= Ready availability of commercial starting materials
= Avoidance of chromatographic separation and purification steps
= Final processing via crystallization
= Final adjustment of the polymorphic modification using class 3 solvents
(in accordance with
FDA guidelines)
Remarkably, a process could be established that meets all of the requirements
mentioned above.
This invention describes the preparation of compound (I) via a surprisingly
highly selective alkylation
on N2 as key step:
I
F
O- -
'NiN-\ ( o
o
(I)
Preparations of N2-substituted indazoles have been described in the
literature, e.g. M.-H. Lin, H.-J.
Liu, W.-C. Lin, C.-K. Kuo, T.-H. Chuang, Org. Biomol. Chem. 2015, /3, 11376.
These procedures,
however, have considerable disadvantages rendering them unsuitable for
technical scale. It is
possible to selectively prepare N2-substituted indazoles via complex sequences
of synthetic steps,
which involve no direct alkylation step. These sequences, however, are long
and tedious and involve
considerable losses ultimately resulting in a low total yield. Therefore,
synthetic routes which allow a
direct preparation of N2-substituted indazoles from 1H-indazole precursors via
direct and selective
alkylation at N2 are most interesting. At the attempt of directly alkylating
the 1H-indazole precursor
of the generic formula (II) generally a mixture made up of the Ni- (Ill) and
N2-alkylated (la)
regioisomers is obtained.
R3 R3 R3
r 1 1 , 1
R2 NO
R2 N....¨......r.0
R2 N....¨....f0
HN HN HN
\ _____________________________ ,.. \ + ---
N,N
N,N
N¨R1
¨N,
HO H HO iRi HO
(II) (III) (Ia)
8

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Indazole and its derivatives, a typical class of aromatic N-heterocycles, have
sparked significant
interest in synthetic and medicinal chemistry due to their diverse biological
activities. Furthermore,
diverse heterocyclic structures could be accessed from indazole-derived N-
heterocyclic carbenes.
Among indazoles, N1/N2-substituted indazoles are widely used as anticancer,
anti-inflammatory,
anti-HIV, and antimicrobial drugs. Generally, the synthesis of N2-substituted
indazoles involves
cyclization procedures from miscellaneous starting materials. Unfortunately,
general methodologies
remain scarce in the literature. Therein, only moderate yields were obtained.
With respect to the current state of technology, several publications are
known and will be discussed
in the following section. None of the published procedures feature reaction
conditions that lead to a
direct N2-selective alkylation using a highly functionalized indazole of type
(II) along with an alkyl
tosylate or halide bearing an alcoholic group of type (IV) as alkylating
agent.
0
40 ii
S ¨0 RI
8 -\ ____________________________________________ ( OH
R2
(IV)
The selectivities and/or yields are low. The problem of the prior art
procedures consists in the limited
functional group tolerance. Thus, only relatively simple alkylating agents
bearing no labile and/or
reactive functional groups apart from the leaving group are used. These agents
are mostly attached
to the respective 1H-indazole via nucleophilic substitution of their halides,
triflates, tosylates, or
mesylates. When more functionalized moieties are used, yield and selectivity
decrease dramatically.
In the following section, the reasons are presented why these prior art
procedures are not applicable
to the challenge at hand:
1. WO 2011/043479: The reactions are carried out in THF at reflux. This does
not work for the
case at hand (alkylating agents of type (IV)). The preparation of the
corresponding triflate
from e.g. the alcohol is not possible, as its decomposition occurs instantly.
In addition, only a
simple substrate with no functionality in the side-chain was used.
2. S. R. Baddam, N. U. Kumar, A. P. Reddy, R. Bandichhor, Tetrahedron Lett.
2013, 54, 1661:
Only simple indazoles without functional groups were used in the reaction.
Only methyl
trichloroacetimidate was used as alkylating agent. Attempts to transfer acid-
catalyzed
conditions to selective alkylation using a functionalized alcoholic alkylating
agent as depicted
9

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
by (IV) at position 2 of an indazole core structure failed. This procedure
cannot easily be
scaled up.
3. Q. Tian, Z. Cheng, H. H. Yajima, S. J. Savage, K. L. Green, T. Humphries,
M. E. Reynolds, S.
Babu, F. Gosselin, D. Askin, Org. Process Res. Dev. 2013, 17, 97: The
preparation of a THP-
ether with preference for N2 of the indazole is presented. This reaction
proceeds via a
different mechanism and does not represent a general method, since the THP-
ether product
cannot be easily converted further. Furthermore, selective methods for
protection of
indazoles using p-methoxybenzyl derivatives under acidic conditions are
presented. Attempts
to transfer these conditions to selective alkylation using a functionalized
alcoholic alkylating
agent as depicted by (IV) at position 2 of an indazole core structure failed.
4. D. J. Slade, N. F. Pelz, W. Bodnar, J. W. Lampe, P. S. Watson, J. Org.
Chem. 2009, 74, 6331:
THP-ether and PMB-protection using acidic conditions (PPTS: pyridinium para-
toluenesulfonate); attempts to transfer these conditions to selective
alkylation using a
functionalized alcoholic alkylating agent as depicted by (IV) at position 2 of
an indazole core
structure failed.
5. M. Cheung, A. Boloor, J. A. Stafford, J. Org. Chem. 2003, 68, 4093: Highly
reactive and highly
carcinogenic Meerwein-salts were used as alkylating agents. This method only
comprises
simple non-functionalized ethyl and methyl Meerwein salts. The reaction
proceeds in polar
ethyl acetate at ambient temperature. These conditions cannot be transferred
to selective
alkylation using a functionalized alcoholic alkylating agent as depicted by
(IV) at position 2 of
an indazole core structure.
ArO Ar, Ary0
HI= X HN
""=-="--NTG HiNx7- j¨FG
'N
4
FG FG FG
dewed undesired L\
FG
Scheme 1: N-alkylation of 1H-indazoles

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Et3OPF6
Et0Ac, rt
\ 1110 14111
JOC 2003, 4093
0 Br 1111 10)8 r
N N
p-Ts0H 1.1
CH2Cl2 H 2
p-Ts0H: p-toluenesulfonic acid PPTS: Pyridin i LIM p-
toluenesulfonate
NH
Br 40 PMFOH Br -
N PMBO CCI3 Br
¨\N¨PMB
< '
inviB tdue H CH2C12
110 -C
PMB: p-methoxybenzyl
JOC 2009, 6331
F- OTf
.4( CyiN 2 ===--,
R2N THF R2N
PCT Int. Appl., 2011043479
Scheme 2: N-alkylation methods of indazoles known from prior art
6. M.-H. Lin, H.-J. Liu, W.-C. Lin, C.-K. Kuo, T.-H. Chuang, Org. Biomol.
Chem. 2015, /3, 11376:
The procedure is N2-selective, however, it cannot be scaled up with Ga and Al
metal used in
stoichiometric amounts. Under the described reaction conditions Broensted
acids are formed
which react with the corresponding metals to give hydrogen gas. Only
relatively simple
substrates are used as alkylating agents. When more functionalized substrates
were used, a
significant decrease in yield was observed. Attempts to transfer these
conditions to selective
alkylation using a functionalized alcoholic alkylating agent as depicted by
(IV) at position 2 of
an indazole core structure failed.
7. G. Luo, L. Chen, G. Dubowchick, J. Org. Chem. 2006, 7/, 5392: 2-
(Trimethylsilyl)ethoxymethyl
chloride (SEM-CI) in THE was used for substitution on N2 of indazoles.
Attempts to transfer
these conditions to selective alkylation using a functionalized alcoholic
alkylating agent as
depicted by (IV) at position 2 of an indazole core structure failed. The
corresponding
products described in this publication are ethers and are not related to our
target molecule.
The use of highly carcinogenic 2-(trimethylsilyl)ethoxymethyl chloride (SEM-
CI) as well as
11

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
benzyloxymethyl chloride (B0M-C1) does not represent a scalable option for
obtaining the
target compound.
8. A. E. Shumeiko, A. A. Afon'kin, N. G. Pazumova, M. L. Kostrikin, Russ. J.
Org. Chem. 2006, 42,
294: Only very simple substrates were used in this method. No significant
selectivity is
reported. A slight preference for N1-alkylation at the indazole was observed.
9. G. A. Jaffari, A. J. Nunn, J. Chem. Soc. Perkin 1 1973, 2371: Very simple
substrates and only
methylation agents were used. A more complex substrate as e.g. a combination
of
formaldehyde with protonated methanol resulted in only N1-substituted product
(ether).
10. V. G. Tsypin et al., Russ. J. Org. Chem. 2002, 38, 90: The reaction
proceeds in sulfuric acid and
chloroform. These conditions cannot be transferred to 2-substituted indazoles.
Only
conversions of simple indazoles with adamanthyl alcohol as sole alkylating
agent are
described.
11. S. K. Jains et al. RSC Advances 2012, 2, 8929: This publication contains
an example of N-
benzylation of indazoles with low selectivity towards N1-substitution. This
KFIalumina-
catalyzed method cannot be applied to 2-substituted indazoles. Attempts to
transfer these
conditions to selective alkylation using a functionalized alcoholic alkylating
agent as depicted
by (IV) at position 2 of an indazole core structure failed.
12. L. Gavara et al. Tetrahedron 2011, 67, 1633 : Only relatively simple
substrates were used. The
described acidic THP-ether formation and benzylation in refluxing THE are not
applicable to
our substrate. Attempts to transfer these conditions to selective alkylation
using a
functionalized alcoholic alkylating agent as depicted by (IV) at position 2 of
an indazole core
structure failed.
13. M. Chakrabarty et al. Tetrahedron 2008, 64, 6711: N2-alkylation was
observed but N1-
alkylated product was obtained preferentially. The described conditions of
using aqueous
sodium hydroxide and phase transfer catalyst in THE are not suitable to
achieve selective
alkylation at position 2 of 1H-indazoles. Attempts to transfer these
conditions to our system
(IV)/(11) failed.
14. M. T. Reddy et al. Der Pharma Chemica 2014, 6, 411: The reaction proceeds
in the
corresponding alkylating agent as solvent. Only the use of highly reactive
ethyl bromoacetate
as alkylating agent is reported. There are no data on the selectivity. These
conditions are not
applicable to compounds as 2-indazoles. Attempts to transfer these conditions
to selective
12

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
alkylation using a functionalized alcoholic alkylating agent as depicted by
(IV) at position 2 of
an indazole core structure failed.
15. S. N. Haydar et al. J. Med. Chem. 2010, 53, 2521: Only simple non-
functionalized alkyl groups
are described (methyl, isopropyl, isobutyl). Cesium carbonate was used as base
and the
reaction resulted in a mixture of Ni- and N2-alkylated products. These
conditions are not are
not suitable to achieve selective alkylation at position 2 of 1H-indazoles.
Attempts to
transfer these conditions to selective alkylation using a functionalized
alcoholic alkylating
agent as depicted by (IV) at position 2 of an indazole core structure failed.
16. Zh. V. Chirkova et al. Russ. J. Org. Chem. 2012, 48, 1557: In this method,
relatively simple
substrates are converted with potassium carbonate as base in DMF. Mixtures of
Ni- and N2-
alkylated products are obtained. The conditions are not are no tsuitable to
achieve selective
alkylation at position 2 of 1H-indazoles.. Attempts to transfer these
conditions to selective
alkylation using a functionalized alcoholic alkylating agent as depicted by
(IV) at position 2 of
an indazole core structure failed.
17. C. Marminon et al. Tetrahedron 2007, 63, 735 : The ortho-substituent R in
position 7 at the
indazole directs the alkylation towards N2 via shielding Ni from electrophilic
attacks. The
conditions, sodium hydride as base in THE, are not suitable to achieve
selective alkylation at
position 2 of 1H-indazoles and preferentially result in alkylation at Ni in
absence of a
substituent in position 7 of the indazole. Attempts to transfer these
conditions to selective
alkylation using a functionalized alcoholic alkylating agent as depicted by
(IV) at position 2 of
an indazole core structure failed.
18. D. A. Nicewicz et al. Angew. Chem. Int. Ed. 2014, 53, 6198: Only simple
substrates were used.
This method describes a photochemical reaction that cannot easily be scaled up
and is not
applicable to a general selective, direct alkylation of 1H-indazoles at
position 2. Very specific
styrene derivatives are used under radical reaction conditions. Attempts to
transfer these
conditions to selective alkylation using a functionalized alcoholic alkylating
agent as depicted
by (IV) at position 2 of an indazole core structure failed.
19. Togni et al. Angew. Chem. Int. Ed. 2011, 50, 1059: This publication solely
describes a special
type of substituent (hypervalent iodine as trifluoromethylation reagent in
combination with
acetonitrile). This special case is not general and cannot be applied to the
synthesis of N2-
alkylated indazoles of type (la) or (Va).
13

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
20. L. Salerno et al. European J. Med. Chem. 2012, 49, 118: This publication
describes the
conversion of indazoles in an a¨bromoketone melt. The reaction conditions
cannot be
transferred to the direct and selective synthesis of N2-alkylated indazoles of
type (I).
Attempts to transfer these conditions to selective alkylation using a
functionalized alcoholic
alkylating agent as depicted by (IV) at position 2 of an indazole core
structure failed.
21. K. W. Hunt, D. A. Moreno, N. Suiter, C. T. Clark, G. Kim, Org. Lett. 2009,
//, 5054: This
publication essentially describes an N1-selective alkylation method with
addition of different
bases. Simple substrates were used. Attempts to transfer these conditions to
selective
alkylation using a functionalized alcoholic alkylating agent as depicted by
(IV) at position 2 of
an indazole core structure failed.
22. J. Yang et al. Synthesis 2016, 48, 1139: This publication describes an N1-
selective base-
catalyzed aza-Michael reaction. No substitution at N2 was observed. Attempts
to transfer
these conditions to selective alkylation using a functionalized alcoholic
alkylating agent as
depicted by (IV) at position 2 of an indazole core structure failed.
23. P. R. Kym et al. J. Med. Chem. 2006, 49, 2339: Essentially N1-alkylations
are described.
Attempts to transfer these conditions to selective alkylation using a
functionalized alcoholic
alkylating agent as depicted by (IV) at position 2 of an indazole core
structure failed.
24. A. J. Souers et al. J. Med. Chem. 2005, 48, 1318: This publication
describes the use of
potassium carbonate as base. This method proceeds mainly with preference for
substitution
at Ni and is therefore not suitable to achieve selective alkylation at
position 2 of 1H-
indazoles. Attempts to transfer these conditions to selective alkylation using
a functionalized
alcoholic alkylating agent as depicted by (IV) at position 2 of an indazole
core structure failed.
25. P. Bethanamudi et al. E-Joumal of Chemistry 2012, 9, 1676: The use of
ionic liquids along
with potassium carbonate as base results in mixtures of Ni- and N2-alkylated
indazoles with
low yields. The selectivity shows a tendency towards substitution at Ni. The
use of ionic
liquid cannot be transferred to our system. Attempts to transfer these
conditions to selective
alkylation using a functionalized alcoholic alkylating agent as depicted by
(IV) at position 2 of
an indazole core structure failed.
26. S. Palit et al. Synthesis 2015, 3371 : The reaction described herein is
essentially non-selective
with a slight preference of substitution at Ni of the indazole. Only simple,
non-functionalized
alkyl groups were used. Sodium hydride and similarly strong bases were used.
Attempts to
14

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
transfer these conditions to selective alkylation using a functionalized
alcoholic alkylating
agent as depicted by (IV) at position 2 of an indazole core structure failed.
It was shown that the compound of formula (I) as well as its precursor (V) can
be synthesized
analogously to methods previously published in the literature via e.g. direct
alkylation with 4-bromo-
2-methylbutan-2-ol using potassium carbonate as base along with potassium
iodide in DMF.
I
F>rN 0
F
F HN
N
-\
0 ....... ,
N ( OH
0
(V)
However, a mixture of Ni- and N2-alkylated products was obtained with a
preference for the N1-
regioisomer (N1:N2 = ca. 2:1). Desired N2-alkylated indazole (V) could also be
obtained in a low yield
as described in W02016/083433, published after the priority date of the
present application, as
described in the following reaction procedure:
930 mg (2.55 mmol) of methyl 5-({[6-(trifluoromethyppyridin-2-
yl]carbonyllamino)-1H-indazole-6-
carboxylate (Vila), 1.06 g of potassium carbonate and 212 mg of potassium
iodide were initially
charged in 9 ml of DMF and the mixture was stirred for 15 min. Then 0.62 ml of
4-bromo-2-
methylbutan-2-ol was added and the mixture was stirred at 60 C overnight. The
mixture was mixed
with water and extracted twice with ethyl acetate, and the extract was washed
three times with
saturated sodium chloride solution, filtered and concentrated. Column
chromatography purification
on silica gel (hexane/ethyl acetate) gave 424 mg (37 %) of the title compound
(V).
Desired N2-alkylated indazole of formula (I) was obtained in an even lower
yield from (11a), as
described in the following reaction procedure:
A mixture of 500 mg (1.37 mmol) of N46-(2-hydroxypropan-2-y1)-1H-indazol-5-y1]-
6-
(trifluoromethyl)pyridine-2-carboxamide (11a), 569 mg of potassium carbonate
and 114 mg of
potassium iodide in 5 ml of DMF was stirred at room temperature for 15 min.
344 mg (1.5
equivalents) of 4-bromo-2-methylbutan-2-ol were added and the mixture was
heated to 100 C for
2 h. Another 0.5 equiv. of 4-bromo-2-methylbutan-2-ol was added and the
mixture was stirred at

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
room temperature overnight. The mixture was mixed with water and extracted
twice with ethyl
acetate, and the combined organic phases were washed with saturated sodium
chloride solution and
filtered through a hydrophobic filter and concentrated. The residue was
purified by column
chromatography purification on silica gel (hexane/ethyl acetate). This gave
100 mg of a product
.. fraction which was stirred with diethyl ether. The solid was filtered and
dried. 60 mg of the title
compound (I) were obtained. Total yield: 160 mg (26 %).
Consumptive preparative HPLC proved indispensable for an efficient separation
of the N1-/N2-
regioismers. This new inventive process aims at an increase in the efficiency
of the synthesis for
scale-up and at a facilitation of the purifications of (I) and (V) via
achieving better selectivity in the
alkylation reactions in favour of substitution at N2 as well as at
establishing a safe process for the
production and handling of 3-hydroxy-3-methylbutyl 4-methylbenzenesulfonate
(VI), which is prone
to decomposition at higher temperatures and under the influence of acid and
base. Also, highly
flammable solvents, such as diethyl ether, which are not suitable for large
scale preparations must be
avoided.
0
41 g -0
(VI)
The present invention provides a process for preparing compounds of the
general formula (la) from
either direct N2-selective alkylation of compounds of the general formula (II)
or via N2-selective
alkylation of compounds of the general formula (VII) resulting in
intermediates of the general
formula (Va) which are converted in a final synthetic step to compounds of the
general formula (la)
via addition of methylmagnesium halide.
16

CA 03022327 2018-10-26
WO 2017/186693 PCT/EP2017/059748
R3 R3
. ,; ,....,
R-
s. II
. (---
R2---N- --fp
HN e,õ,..... HN _
-.....-.:---x--\.õ,
7
1 N ____________________________________________ yls, N-R1
111::;1-H HO
(0) (la)
R3 R3 R3
r
ar0 ly,0
,0
R2 k . R2 1-7 HNI--
HN MeMgX HN ,
--- HO ' .. -
-- -
Oy= *,... ---N= " -"N'
H
0 0
(VII) (Va) (la)
in which
R is
/ (OH;
i
R2 is difluoromethyl, trifluoromethyl or methyl; and
R3 is hydrogen, alkyl or fluorine;
X is F, CI, Br or I
with preferably R2 = trifluoromethyl and R3 = H and X =CI:
o
-, -,
S-o¨\
____________________________________ OH I 0
F3C '1\1-- F3CN -
HIV. (VI) HN
-r. ;--
H --1\1N¨\ ( OH
HO HO
(11a) (I)
0
F3C-1\1 0 (y- O OH F3 ---',--NI0
F3C'N 0 'y-
HN (VI) HN MeMgCI HN
i1IIIi\ _,..--,----\-
N __________ .
0 0 - N¨\
N N \ ( OH <OH
H HO 1
0 0
(Vila) (V) (I)
Unexpectedly, we found that the use of 3-hydroxy-3-methylbutyl 4-
methylbenzenesulfonate (VI)
along with N,N-diisopropylethylamine as base in toluene resulted in highly N2-
selective alkylation
reactions for indazoles (V) and (11a). The N2-selectivities in these
alkylation reactions of complexly
functionalized indazoles with an alkyl tosylate bearing a reactive functional
group are unprecedented
and therefore highly inventive. Upon reaction of compounds of the general
formula (II) or (VII) with
3-hydroxy-3-methylbutyl 4-methylbenzenesulfonate (VI) in a hydrocarbon
solvent, such as toluene,
17

CA 03022327 2018-10-26
WO 2017/186693 PCT/EP2017/059748
xylene or chlorobenzene, with addition of an organic base, such as N,N-
diisopropylethylamine or
triethylamine, the desired N2-isomers (I) and (V) are obtained with very high
selectivities.
Surprisingly, the selectivity in the alkylation reaction of (11a) with (VI)
was even higher than that
observed in the alkylation of (Vila).
. 9
,
F,c'N'r 0 KOH
F3CNr F3CNr
. 0 __________________________________
HN (VI) HN HN 0 .
N .. Rip_ p¨\ KOH + N
0 0 0 NuL)H
N N µ ____
H
0 0 0
(Vila) (V) (VIII)
Selectivity: 10 = . 1
0
F3CNC31 0 ,N ________ KOH
F3CN C31 F3CNr
0
HN (VI) HN HN . 0 .
N N KOH NuL)H
HO H HO HO
(11a) (I) (111a)
Selectivity: 26 = . i
Remarkably, the conversion of the starting indazole to the desired N2-
alkylated product was much
higher for (11a) than (Vila). Thus, the HPLC ratios of N2-alkylated product to
starting indazole at the
end of the reaction was only less than 3 : 1 for (V) : (Vila) and 30 : 1 for
(I) : (11a) (HPLC). Interestingly,
we observed that the reaction could be well performed via slow simultaneous
addition of an organic
base and a solution of an alkylating agent in unpolar hydrocarbon solvent,
such as toluene, xylene or
chlorobenzene. It proved beneficial to have a (slight) excess of base at each
time point during the
reaction. Another method works via slow addition of a solution of the
alkylating agent in an unpolar
solvent, such as toluene, xylene or chlorobenzene, to a mixture of the
starting 1H-indazole and an
excess of organic base (N,N-dicyclohexylamine or triethylamine, preferably N,N-
diisopropylethyl-
amine) in the aforementioned solvent (toluene or xylene) at elevated
temperature (>100 C). The
reaction of (Vila) to (V) worked best when 21 equiv. of base (N,N-
dicyclohexylamine or triethylamine,
preferably N,N-diisopropylethylamine) were used. A mixture of indazole (Vila)
and base in toluene
(6.5 volumes) was heated to 100¨ 110 C. In order to ensure a safe process, 5
equiv. of 3-hydroxy-3-
methylbutyl 4-methylbenzenesulfonate (VI) are added to the reaction mixture as
a solution in
1 volume toluene over a period of 10 h. After complete addition, the reaction
is stirred for additional
12 ¨ 18 hours, (preferably 15 hours) at 100¨ 110 C. Optionally, the stirring
time can be 14 - 24 h
(preferably 18 h) at 100 ¨ 110 C as well. Preferably, the reaction mixture is
stirred for 18 h at 110 C.
For the reaction of (Vila) to (V), the conversion stalls at an average ratio
of starting indazole to N2-
alkylated product of 2.8 : 1 (ratio of area% HPLC). Thus, in order to also
regain the non-converted
18

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
starting indazole (Vila), a column chromatography is best performed for
purification of (V).
Remarkably, a column chromatography procedure could be found that allowed the
efficient
purification of (V) to 99.5 area% HPLC and clean isolation of (Vila) on kg-
scale. (V) is obtained with an
overall yield comprising the alkylation and ensuing chromatography step in the
range of 45 - 47 %.
This procedure was performed at kg-scale.
In case of the transformation of (11a) into (I), we found that a high
conversion was achieved when 4.0
equiv. of a 15-35 wt% solution of 3-hydroxy-3-methyl butyl 4-
methylbenzenesulfonate (VI) in toluene
were dosed over 5 - 15 h (preferably 10 h) to a suspension of (11a), 4.8
equiv. of an organic base
(preferably N,N-diisopropylethylamine) and toluene at the reflux temperature
of toluene W.10 C
internal temperature) under ambient pressure. After complete addition, the
reaction is stirred for
h to 24 h (preferably 18 h) in order to reduce the amount of remaining (VI) in
the mixture.
(V) is converted to the target compound (I) via addition of methyl magnesium
halide. The procedure
15 used in the research synthesis of (I) is disclosed in W02016/083433,
published after the priority date
of the present application and described here:
705 mg (1.57 mmol) of methyl 2-(3-hydroxy-3-methylbuty1)-5-({[6-
(trifluoromethyl)pyridine-2-
yl]carbonyllamino)-2H-indazole-6-carboxylate (V) were initially charged in 10
ml of THE and cooled in
an ice-water cooling bath. 2.6 ml (5.0 equiv.) of 3 M methylmagnesium bromide
solution in diethyl
ether were added and the mixture was left to stir while cooling with an ice
bath for 1 h and at room
temperature for 4.5 h. Another 1 equiv. of the methylmagnesium bromide
solution was added and
the mixture was left to stir at room temperature for 20.5 h. Another 1 equiv.
again of the
methylmagnesium bromide solution was added and the mixture was left to stir at
room temperature
for 22 h. The reaction mixture was mixed with saturated aqueous ammonium
chloride solution,
stirred and extracted three times with ethyl acetate. The combined organic
phases were washed with
sodium chloride solution, filtered through a hydrophobic filter and
concentrated. This gave 790 mg of
a residue which was purified by means of preparative HPLC. This gave 234 mg of
the title compound
and 164 mg of a product fraction which was stirred with diethyl ether. After
filtration with suction
followed by drying, a further 146 mg of the title compound were obtained.
Total yield: 398 mg (56 %)
This procedure is not suitable for large scale production due to the following
reasons:
= The use of diethylether must be avoided due to its low ignition point and
its high explosive
potential.
= The relatively costly methylmagnesium bromide was used instead of the
more common
methylmagnesium chloride which is easier to procure.
19

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
= The total time of the reaction is very long (47 h!)
= The reaction is accompanied by the formation of many unwanted side-
products, so that a
preparative HPLC had to be used for purification.
= Chromatographic separations should be avoided on technical scale, as they
usually require
an uneconomical consumption of organic solvents.
= No crystallization procedure has been described. According to the usual
practice in research
laboratories, compound (I) was evaporated to dryness. This operation is not
feasible on
technical scale.
Surprisingly, we found that compound (V) could be prepared with a
significantly higher yield when
methylmagnesium chloride in THE was used instead. The reaction proceeds with
less side-products
which, using the research method as disclosed in W02016/083433, had to be
removed via
preparative HPLC. The reaction was found to proceed best with THE as solvent.
6 equiv.
methylmagnesium chloride (ca. 3 M in THE) are stirred and kept at -10 to -15
C. Within 1-2 h
(preferably 1.75 h) compound (V) is added dropwise to the mixture as a
solution in THE. The reaction
mixture is stirred for 30 min at the indicated temperature. The cold reaction
mixture is subsequently
quenched by being dosed into an aqueous solution of citric acid. The resulting
mixture is stirred
vigorously. Phases are separated. The aqueous phase is extracted with ethyl
acetate. The combined
organic phases are washed with water. A solvent swap to ethanol is performed.
The resulting
solution is warmed to 31 ¨ 32 C and stirred. The crude product is
crystallized by adding water over a
period of 1 h. The resulting suspension is then cooled to 20 C within 1 h and
the crude product is
isolated via filtration and washed with a mixture of ethanol and water. The
crude product is dried.
For purification, the product is subjected to further crystallization using a
mixture of
acetone/toluene 1:9. The crude material is dissolved in this mixture at app.
80 C. The solution is
cooled to 55 C. It proved advantageous to add seeding crystals at this
temperature. The resulting
suspension is further cooled to 20 C within 2 h, the product is filtered off,
washed with a mixture of
acetone/toluene 1:9 and toluene and dried.
In order to receive a defined crystalline form, the product is subjected to
crystallization with ethanol
and water analogously to the procedure described above. Using this procedure,
the desired
compound (I) is obtained with high purity (>97 area% HPLC; >96% content) and
good yields
(55 - 77 %). Remarkably, the yields were higher (72 and 77 %) when the
reaction was run at larger
scale (kg).

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Notably, we found that the alkylation reaction of (11a) to (I) gave the best
results when only 4.5 to
6 equiv. base (N,N-dicyclohexylamine or triethylamine, preferably N,N-
diisopropylethylamine) were
used. We also found that a simultaneous and slow addition of asolution of (VI)
in toluene (15-40
wt%; preferably 25 wt%) proved beneficial. When the addition is performed
simultaneously, a slight
excess of base must be present in the reaction mixture for the alkylation to
proceed best. It is also
possible to slowly add the solution of (VI) in an unpolar hydrocarbon solvent,
in particular toluene, to
a mixture of (11a) and organic base in the same unpolar hydrocarbon solvent.
For this reaction, a
toluene solution of (VI) has been prepared according to an optimized procedure
with respect to
safety and handling, as (VI) is prone to exothermic decomposition. Thus, (11a)
is suspended in toluene
(ca. 6.5 volumes) and heated to 100 - 3.12 C (preferably reflux temperature
of toluene as internal
temperature). After complete addition, the reaction mixture is stirred for 18
h at 100¨ 3.12 C.
After complete addition, the reaction was stirred for 15 to 24 hours,
preferably 18 h, in order to
decrease the amount of the remaining excess of the alkylating agent (VI). The
reaction mixture is
then cooled to a temperature of 40 C and concentrated under vacuum.
.. The reaction mixture is then cooled to 40 C and concentrated. A phase
extraction sequence follows
using ethyl acetate, a mixture of acetic acid/ water, and water. The organic
phase is concentrated
and a solvent swap to isopropanol is performed. The desired product (I) is
crystallized via slow
addition of water. In some cases, it proved useful to seed the mixture with
small amounts of crystals
in order to obtain a reproducible crystallization. After prolonged stirring of
the resulting suspension,
.. the product is isolated via filtration, washed with a mixture of
isopropanol and water, and finally
water. The product is dried at 50-60 C under vacuum resulting typically in
60¨ 90 % yield. The purity
of the crude product typically amounts to 76-89 % (area% HPLC; method D) (70
to 90 wt% content)
with less than 6 % (HPLC) of N1-regioisomer. This work-up, however, proved
difficult at large scale
(1.2 kg), as the content of the product was lower than that originally
obtained at lab scale (down to
61 wt%; 71 area% HPLC; method C; 76 area% HPLC; method D).
The crude product can be purified via repetitive crystallization from a
toluene/acetone mixture
similar to the crystallization procedure applied after the reaction of (V) to
(I). Here, we found it
beneficial to add activated charcoal (0.1 - 0.4 equiv.) in order to achieve
optimal results. (I) is thus
received with purities of 95 to >99 area% HPLC.
The preparation of cGMP material which will also be used in clinical trials
requires additional
purification. In addition, since the active pharmaceutical ingredient will be
used for tablet
production, a procedure is required that reproducibly furnishes the identical
crystalline form.
Surprisingly, a defined crystal form could be installed via recrystallization
with ethanol and water. For
cGMP filtration the compound is first dissolved in ethanol passed through a
particle filter and
21

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
subsequently crystallized via addition of water. The pure product is usually
obtained in 35 - 56% with
high purity and content.
Since the above-described work-up resulted in content fluctuations when
applied at larger scale, we
searched for a more efficient work-up and purification.
Surprisingly, we found that n-butyl acetate proved suitable as solvent for an
efficient purification via
crystallization of crude (0. Therefore, n-butyl acetate was used both as
solvent in the extractive
work-up and as solvent for crystallization. The crystallization was performed
using a warm-cool cycle,
which notably gave material that could be easily handled for filtration. "Warm-
cool cycle" in the
aforementioned sense means, that the crude material was dissolved in n-butyl
acetate at app. 93 C,
kept at this temperature for 1 h, then cooled to 83 C within 30 min. The
material started to
crystallize at this temperature, optionally seeding crystals were added. The
resulting suspension was
stirred for 10 min and then cooled to 60 C within 2 h. At this temperature,
the suspension was
stirred for at least 30 min before it was warmed to 78 C within 30 min. The
mixture was stirred at
this temperature for at least 30 min, before it was cooled to 22 C within 6
h. The resulting
suspension could be easily filtrated. The described warm-cool cycle proved
essential for obtaining
easily filterable material. Using this procedure, compound (I) was received
with high purity (>97
area%) and yields >50 %. This procedure was successfully carried out at 1 kg
and 18 kg scale.
For achieving cGMP (current Good Manufacturing Practice) quality by reducing
the amount of
potentially genotoxic (VI) in the final product (I) to an acceptable level
(<20 ppm) and for obtaining a
defined crystalline form, (I) was dissolved in ethanol at 55 C and the
solution was subjected to
clarification filtration. The solution was then heated to 65 C and water was
added within a time
regimen, which is in analogy to that described by the mathematical equation of
a cubic dosing curve*
(amount water added vs. addition time):
( t )3
= (Mtotal) X Mstarb
LB
whereby
m(t) = amount H20 vs. addition time [kg]
Mtotal = total amount of H20 added via cubic addition [kg]
Mstart = amount of water present before start of cubic addition [kg]
t = time [h]
tB = total addition time [h].
* Principle of cubic dosing curve is described by S. Kim et al. in Org.
Process Res. Dev. 2005, 9, 894.
22

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
The addition of water to a solution of compound (I) in ethanol at 65 C within
the above-described
time regimen ("cubic dosing curve") results in product particles which are
characterized by
significantly larger crystal sizes (see figure 2) and a defined particle size
distribution compared to
product particles obtained after water addition at the same temperature (65
C), but within a time
.. regimen described by the equation of a linear function (y = a x z + b),
i.e. "linear water addition".
After complete addition of the total amount of water and additional stirring
at 65 C, the suspension
was cooled to 20 C. The precipitate was filtered off and washed with a
mixture of water and ethanol
and dried. The resulting crystalline particles have a defined shape and the
desired properties
required for formulation of a pharmaceutical composition, such as a tablet
(see Experimental
Section: XRPD Reflexes) with high purity (>97 area%) and high yield (>90 %).
The novel crystallization procedure provides benefit with regard to filtration
and operative handling
of the crystalline material obtained according to the above-described protocol
("cubic dosing curve").
Thus, crystals obtained via the "cubic dosing curve" crystallization procedure
showed superior
filtration properties, such as a lower amount of residual moisture (wf= 28 %
weight) after filtration, a
.. lower resistance of the filtration cake (a= 2.141.012 m-2) and a
considerably higher volume flow rate
(vF= 12,484 1/m2h) than crystals obtained via the "linear water addition"
crystallization procedure
(wf= 37 % weight; a= 8.6*1012 m-2; vf= 3,306 1/m2h). The a- and vf-values were
determined in a
normalized filtration experiment analogous to the VD! 2762 Part 2 guideline
dated December 2010.
The residual moisture was determined in a drying oven (Heraeus vacutherm, 30
mbar, 50 C,
overnight) and with a Halogen Moisture Anaylzer HG53 (Mettler Toledo) at 120
C.
Additionally, the obtained crystals can be defined by a specific particle size
distribution of x90:
7.7-9.7 m; x50: 2.7-3.2 m; x10: 0.9-1.0 m.
In contrast, crystals obtained with the "linear water addition" are defined by
a particle size
.. distribution of x90: 7.7-9.7 m; x50: 2.7-3.2 m; x10: 0.9-1.0 m.
The most commonly used metrics when describing particle size distributions are
x-values (x10, x50 &
x90) which are the intercepts for 10%, 50% and 90% of the cumulative mass.x-
Values can be thought
of as the diameter of the sphere which divides the samples mass into a
specified percentage when
the particles are arranged on an ascending mass basis. For example, the x10 is
the diameter at which
10% of the sample's mass is comprised of particles with a diameter less than
this value. The x50
represents the diameter of the particle that 50% of a sample's mass is smaller
than and 50% of a
sample's mass is larger than.
This procedure is well compatible with technical scales.
23

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Product that is obtained from this crystallization procedure possesses the
desired properties
required for preparation of a pharmaceutical composition, such as a tablet
(see Experimental
Section: XRPD Reflexes). The crystalline material obtained via the above
described crystallization
procedure displays good stability during storage. It can also be easily
micronized without losing its
crystal properties.
It must be emphasized that the N2-selective alkylation of a complexly
functionalized indazole using
an alkylating agent bearing reactive functionalities apart from the leaving
group is novel, without
precedence in the literature and therefore a scientifically highly significant
invention for the
preparation of such substitution patterns.
In the previous non-selective alkylation reactions, 4-bromo-2-methylbutan-2-ol
(CAS No. 35979-69-2)
was used as alkylating agent. Larger quantities of this material are difficult
to procure so that this
compound does not represent a viable option on scale. We therefore decided to
switch to the
corresponding tosylate (VI) (CAS No. 17689-66-6) which can be prepared from
readily available
3-methylbutane-1,3-diol (IX) (CAS No. 2568-33-4) and p-toluenesulfonyl
chloride (X) (CAS No.
98-59-9).
0
OH
HO
0
OH CI =0
(IX) (X) (VI)
Notably, we found that the reaction can be carried out at a very high
concentration of (IX) in
dichloromethane (total: 5.8 - 6 volumes). (IX) is first mixed with
triethylamine and
4-dimethylaminopyridinee (CAS No. 1122-58-3) in dichloromethane (2 volumes) at
20 - 25 C. This
reaction mixture is cooled to 0 5 C. A solution of (X) in dichloromethane (2 -
2.1 volumes) is added
over a period of 75 - 90 min. The reaction is warmed to ambient temperature
(20 - 25 C) and stirred
for 12 - 18 h (preferably 15 h). The reaction mixture is quenched with water.
The pH is adjusted to
1.5 - 2. Phases are separated. Half-saturated aq. NaCI-solution is added to
the organic phase and the
pH is adjusted to 7 - 7.5 using saturated aq. NaHCO3-solution. Phases are
separated and the organic
phase is concentrated using a rotary evaporator. At technical scale (1.5 kg of
starting material (IX))
repeatedly defined amounts of dichloromethane are added to the residue and
evaporated in order
to remove remaining water. The compound was obtained as a slightly yellow to
colorless viscous oil
in yields from 90 - 98 % and a purity of typically around 90 area% HPLC.
24

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Remarkably, DSC measurements on (VI) showed that the compound is prone to
exothermic
decomposition at around 100 C. Acids and additives such as rust were shown to
promote this
decomposition. Therefore, a more safe and straightforward process for the
preparation of (VI) had to
be found. Surprisingly, we discovered that (VI) can be directly prepared as a
concentrated solution
(15-40 wt%) in toluene at low temperature. Thus, (IX) is emulsified in 1.5
volumes toluene. The
mixture is cooled to 0 C and 1.1 equiv. triethylamine is added followed by
0.05 equiv.
4-dimethylaminopyridinee. A highly concentrated solution of (X) in toluene
(1.6 volumes) is dropped
to the reaction mixture at 0 C over a period of 2 h. Stirring is continued
for 12 - 18 h (preferably 15
h) at 0 C. The precipitate (triethylammonium chloride) is filtered off and a
clear solution of (IV) in
toluene is obtained. Remarkably, this solution can directly be used in the N2-
selective alkylation
reaction without any further work-up or purification. This procedure avoids
the exposure of (VI) to
heat, acid and large excess of base. Since the toluene solution of (VI) is
telescoped and used directly
after filtration in the N2-selective alkylation reaction of (11a) to (I), it
proved crucial to for the final
purity of (I) to meet the cGMP purity requirements that a slight excess of 3-
methylbutane-1,3-diol
(IX) towards p-toluenesulfonyl chloride (X) is used in the production of the
solution of (VI) and to
make sure that only very small amounts of (X) (<0.05 area%, HPLC) are still
present in the solution. In
order to have the best possible control over the stoichiometries of (IX) vs.
(X), it is beneficial to
subject the relative hygroscopic compound (IX) in a first step to an
azeotropic distillation with
toluene in order to remove water.
The preparations of compounds with the general formula (II) are described in
WO 2015/091426. This
new inventive process focuses on the compound shown by formula (11a):
I
F3C Nr
HN
\
N
,
N
HO H
(Ha)
In the published patent application WO 2015/091426, compound (11a) is
described to be prepared via
reaction of the methyl ester (Vila) with a solution of methylmagnesium bromide
in diethylether.

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
I
F3C Nr
HN
\ 0 N
,
N
H
0
(Vila)
After work-up, the crude product is subjected to a column chromatographic
purification furnishing
compound (11a) in 45 % yield.
This procedure is not suitable for a production of (11a) on technical scale
due to the following
drawbacks:
= The use of diethylether must be avoided due to its low ignition point and
its high explosive
potential.
= The relatively costly methylmagnesium bromide was used instead of the
more common
methylmagnesium chloride which is easier to procure.
= Chromatographic separations should be avoided on technical scale as they
usually require a
massive uneconomical consumption of organic solvents.
= No crystallization procedure has been described. According to the usual
practice in research
laboratories, compound (11a) was evaporated until dryness. This operation is
not feasible on
technical scale.
Surprisingly, it was found that compound (11a) could be prepared with a
significantly higher yield
when methylmagnesium chloride and lithium chloride (2:1) in THE were used
instead. The reactions
proceeded with less side-products which, using the old method described in WO
2015/091426, had
to be removed via tedious column chromatography. The reaction was found to
proceed best with
THE as solvent. 6-10 equiv. methylmagnesium chloride (ca. 3 M in THE) and 3-5
equiv. lithium
chloride are stirred and kept at -10 to 0 C. Within 1 - 3 h (preferably 2 h)
compound (Vila) is dropped
to the mixture as a solution in THE. The reaction mixture is stirred for 5 to
30 min at the indicated
temperature and subsequently quenched by being poured into water. The
resulting mixture is stirred
vigorously. The pH of the mixture is then adjusted to ca. 4.0 via addition of
a mineral or organic acid
(preferably citric acid) and ethyl acetate is added. Phases were separated and
the organic phase was
washed several times with brine (aqueous sodium chloride solution). The
resulting organic solution
was subjected to a solvent swap with toluene via distillation. During this
process, compound (11a)
started to crystallize and could be isolated via filtration. The precipitate
was dried at elevated
26

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
temperature (50-60 C) under vacuum. Typically, yields at this stage were in
the range of 80-96 % and
purities between 95-99 area% HPLC; method A, see experimental).
For the preparation of cGMP material it proved advantageous to finally stir
this product in a mixture
of isopropanol/water (1:1; 2 to 10 volumes relative to input material). The
material is stirred for
1 - 5 h, preferably 3 h. It is then filtrated and washed twice with small
amounts of a 1:1
isopropanol/water mixture. The product is dried at elevated temperature (50 -
60 C) under vacuum.
Typically, yields >90 % and purities >97 area% (H PLC; method A) are achieved.
In the following examples in the experimental section, a variant (see example
#2, variant #3) is also
described in which, after treatment with activated charcoal, a solvent swap
directly to isopropanol is
performed. The product is crystallized by addition of water. In this way, the
product is directly
obtained with very high purity.
The preparation of compound (Vila) has also been described in the patent
application
WO 2015/091426. Thereby, 6-(trifluoromethyl)pyridinee-2-carboxylic acid (XI)
(CAS No.: 21190-87-4)
was coupled with aniline (XII) (methyl-5-amino-1H-indazol-6-carboxylate; CAS
No.: 1000373-79-4)
using 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridineium 3-
oxid hexafluoro-
phosphate (CAS No.: 148893-10-1) as coupling reagent. Amide (Vila) was
obtained with 84 % yield.
, I
+ H2N F3C/N 0
_________________________ \ N 11.. F3CN HN OH 0
NI \
N H
0 0 0 NI
(XI) (XII) H
0
(Vila)
Due to safety reasons, an up-scaling of uronium-based coupling reagents is not
possible for the
reasons of its explosive potential. Therefore, an alternative coupling method
had to be found. The
safe and scalable method for the preparation of amide (Vila) is based on the
use of T3P (2,4,6-
tripropy1-1,3,5,2,4,6-trioxatriphosphorinane-2,4,6-trioxide; CAS No.: 68957-94-
8) as coupling reagent.
The reaction proceeds smoothly and furnishes amide (Vila) with high yields. In
a one-pot process,
carboxylic acid (XI) (best used with a slight shortage of (XI) relative to
aniline (XII) ,ca. 0.90-0.95
equiv.) is placed along with 1.5 equiv. N,N-diisopropylethylamine in 7-16
volumes THF. Subsequently,
27

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
2 equiv. T3P (50 wt% solution in ethyl acetate) are slowly added at 0 - 5 C
over a period of 45 min.
The reaction mixture is additionally stirred for 2 -4 h (preferably 2 h) at 0 -
5 C.
The cold mixture was then quenched with (cold) water, its pH adjusted with
sodium carbonate aq.
solution or alternatively ammonium hydroxide solution to 7.5. The resulting
suspension was then
(when only 7 volumes of THE were used for the reaction) warmed to ambient
temperature and
filtered. The product was washed with water and ethanol and dried under vacuum
at 45 C. In case of
16 volumes of THE, the THE/ethyl acetate mixture was largely distilled off
(200 mbar, 45-50 C
internal temperature). Subsequently, water and ethanol were added and the pH
was adjusted to 7.0
by adding sodium carbonate aq. solution. The mixture was stirred 1-5 h,
preferably 1-2 h, at 50 C,
then cooled to 20 - 25 C and stirred for 10 - 30 min. The product was
isolated via filtration and
subsequently washed with a mixture of ethanol and water and finally dried
under vacuum at 45 C.
With this process, typically high yields between 84-96 % were obtained. The
purity was in all cases
>98 area% (HPLC; methods A & B).
In some cases, especially when aniline (XII) of poor optical quality (e.g.
dark brown color) was used as
starting material, it proved useful to perform a treatment with activated
charcoal. This procedure is
described in the following section:
Crude amide (Vila) was dissolved in a mixture of methanol and THE (2:1) and
activated charcoal was
added. The mixture was heated to 60 - 65 C for 1 - 1.5 h. The activated
charcoal was filtered off and
the filtrate was concentrated (down to 2 volumes relative to input material).
Water was added and
the product precipitated, was filtered, washed and dried at 55 - 60 C (under
vacuum).
Compounds (XI) and (XII) have been reported in the literature and both are
commercially available in
large quantities.
XI: Cottet, Fabrice; Marull, Marc; Lefebvre, Olivier; Schlosser, Manfred,
European Journal of Organic
Chemistry, 2003, 8 p. 1559 ¨ 1568; Carter, Percy H.; Cherney, Robert J.; Batt,
Douglas G.; Duncia,
John V.; Gardner, Daniel S.; Ko, Soo S.; Srivastava, Anurag S.; Yang, Michael
G. Patent:
U52005/54627 Al, 2005; Ashimori; Ono; Uchida; Ohtaki; Fukaya; Watanabe;
Yokoyama Chemical
and Pharmaceutical Bulletin, 1990, vol. 38, 9 p. 2446 - 2458
XII: Nissan Chemical Industries, Ltd.; CHUGAI SEIYAKU KABUSHIKI KAISHA ,
EP2045253 Al, 2009.
Evaluation of the total processes:
The following schemes depict the total syntheses of pure product (I) from
aniline (XII). When
calculating with the best yields achieved for each step, a total average yield
of approximately 35 % is
28

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
obtained for the route via N2-selective preparation of (V). This also includes
the installation of the
final crystalline form.
H2N 40
\,N
0
.-- N
H
= H 0
(XII)
X) "1"3P
ar-k, 0
Et0AciTH1 0 c
"NCI (X). Et3N 80-96%
OH
DtvlAn ; oat.) V VI)
CH C to rt,
jO ut.e 98% F3c- i.
F3C N 1H HN 0
\
I '. ah ___ i ( OH Ts0"¨"' i vi }
N--1 õ...0 N
0
--- WI "N' (f. - It. .) H
DIPEt atuene, rf 0 (Vila)
6 crude M
chromate hy
chromasil 1S: = -1) A
gradient n-he. , = ',DAG
45-4,
i (over 2 steps)
,--- F3C............N,..kr0 MeMgCI (F quiv.), F3C1 0 N
74-, ' - = ,
i ( OH _ ¨sii. HN µ ( OH
HNYa---\-- N--/ .õ..-
fecryst with Et011/H20; N-1
..õ0,...õ ,... ---N=
11 pure (V) then with acetoneltoluene;
then with Et01-111-Ii0
0 (I)
55-77%
The synthetic route via (11a) completely avoids column chromatographic
purification and furnishes
the desired compound (1) with very high purity (>98 area%; method C) and
defined crystalline needle
form and size (see figure 2). The total yield is higher than that obtained
after using the synthetic
route via (V): total average yield of approximately 42%.
jell
Ho.,--,
TsC
E ' 66%
Phi. T
.,.. 1
..+C ..,f
F3C ''N 4 F3C(.1 ro 1) Tse r r ).0N
A
r , '
1:1211ir MN HN J
%
0 I IS N' S ....N14
.-= =
.1 HO H B. . . MO
o
(1/11a) EtC ilq. 81.10A = , - I. (0
(Ha) Bi =
Ett.... .4aCi.
r r V 2) EtC. 35 *C recryst.
ises. ,0 50-60%
7t- /0
29

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
When comparing these total yields with the published prior art data with
regard to
1. amide coupling (preparation of VI): 84 % yield;
2. Grignard reaction followed by chromatographic purification: Grignard
reaction on (Vila):
45 % yield; on (V): 56 % yield.
3. alkylation with 4-bromo-2-methylbutan-2-ol analogously to methods known to
the skilled
person followed by chromatographic purification: alkylation of (Vila): 37 %
yield; alkylation of
(11a): 26 % yield,
the advantages of the new processes become very clear:
With the prior method a total yield of only 9.8 - 17.4 % could be achieved
with an installation of the
final crystalline needle form not included.
To conclude, the new inventive processes furnish compound (I) with 2.4 (route
via (V)) to 4.3 times
(route via (11a)) higher total yields as compared to the prior art. They,
moreover, include the directed
and reproducible preparation of a defined crystalline needle form and size
(see figure 2).
Hence, in a first aspect, the present invention relates to a method of
preparing a compound of
formula (I) via the following steps shown in reaction scheme IA, infra :
c,C)e
X 0
F3c r 0 F3
MeMgCI
=
0 H.
(Vila)
(11a)
jOH
Ts
(VI)
in
aromatic hydrocarbon solvent
V
cX)r 0
F3
H ( OH
W-N1'
HO
Scheme IA.

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
In an embodiment of the first aspect, the present invention relates to a
method of preparing a
compound of formula (I) via the following steps shown in reaction scheme!,
infra:
F3C
,ae F3C
o o -rscrj H F3c..--01 0
N N
HN 0 N MeMgC1 HN is (VI) HN ...., / ( OH
N ¨a N toluene, N¨=
N
= N LiCI N
N,N-
H H = H H=
0 diisopropylethylamine
(Vila) (I)
(11a)
Scheme I.
In an embodiment of the first aspect, the present invention relates to a
method of preparing a
compound of formula (I):
I
F3c i\r.r0
HOH 0..., N j ( OH
N
(I)
comprising the following step (A):
wherein a compound of formula (11a):
, I
F3C1\,
H
\ ,N
N
H H
(11a)
is allowed to react with a compound of formula (VI):
31

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
ccOH
Ts
(VI)
,
optionally in the presence of an organic base, particularly a weak base, such
as a tertiary amine, such
as N,N-diisopropylethylamine for example,
optionally in an aromatic hydrocarbon solvent, such as toluene, xylene and
mesitylene for example,
thereby providing said compound of formula (I).
In an embodiment of the first aspect, the present invention relates to a
method of preparing a
compound of formula (I) as described supra, wherein said aromatic hydrocarbon
solvent is toluene.
In an embodiment of the first aspect, the present invention relates to a
method of preparing a
compound of formula (I) as described supra, wherein said organic base is N,N-
diisopropylethylamine.
In an embodiment of the first aspect, the present invention relates to a
method of preparing a
compound of formula (I) as described supra, wherein said compound of formula
(11a) :
, I
F3C1\,
H
\ ,N
N
H H
(11a)
is prepared by the following step (B) :
wherein a compound of formula (Vila) :
32

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
F3c,Nr0
,N
0
(Vila)
is allowed to react with a reductive methylating agent, such as a
methylmetallic agent, such as a
methylmagnesium halide, such as methylmagnesium chloride for example,
optionally in the presence of an alkali metal halide, such as lithium chloride
for example,
thereby providing said compound of formula (11a).
In an embodiment of the first aspect, the present invention relates to a
method of preparing a
compound of formula (I) as described supra, wherein said compound of formula
(Vila) :
1
HL-
F3cNr0
,N
0
(Vila)
is prepared by the following step (C) :
wherein a compound of formula (XII) :
0
is allowed to react with a compound of formula (IX) :
33

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
(XI)
optionally in the presence of an organic base, particularly a weak organic
base, such as a tertiary
amine, such as N,N-diisopropylethylamine for example,
optionally in the presence of a coupling agent, such as 2,4,6-tripropy1-
1,3,5,2,4,6-
trioxatriphosphinane 2,4,6-trioxide (T3P) for example,
thereby providing said compound of formula (Vila).
In an embodiment of the first aspect, the present invention relates to a
method of preparing a
compound of formula (I) as described supra, wherein said compound of formula
(I) is purified by
crystallization, particularly from a solvent or a mixture of solvents such as
a mixture of acetone and
toluene, optionally in the presence of activated charcoal,
optionally followed by a further crystallization from a solvent such as
ethanol for example.
In an embodiment of the first aspect, the present invention relates to a
method of preparing a
compound of formula (I) as described supra, wherein said compound of formula
(I) is in the form of
crystalline needles (A) which corresponds to a hydrate form of compound of
formula (I).
In accordance with a second aspect, the present invention relates to
crystalline needles (A) of the
compound of formula (I) which corresponds to a hydrate form of compound of
formula (I).:
I
F3c. Nr 0
( OH
Nfri
1\11
HO
II)
as prepared by the method as described supra.
In accordance with a third aspect, the present invention relates to
crystalline needles (A) of the
compound of formula (I) which corresponds to a hydrate form of compound of
formula (I):
34

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
n
F3c i\r'r0
H N j ( OH
W1\11
HO
(I) .
In accordance with an embodiment of the third aspect, the present invention
relates to said
crystalline needles (A) which corresponds to a hydrate form of compound of
formula (I) as described
supra, having an XRPD peak maxima [ 2Theta] (Copper (Cu)) as follows:
Table 1: XRPD of crystalline needles (A) which correspond to hydrate form of
compound (I)
Reflection [Peakmaxima 2Theta]
Hydrat
6,2
7,9
9,4
10,8
12,5
13,0
13,8
15,0
15,3
15,5
15,7
16,0
16,3
17,0
18,0
18,2
18,7
19,3
20,1
20,3

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Reflection [Peakmaxima 2Theta]
Hydrat
20,8
21,0
21,4
21,7
22,9
23,4
24,0
24,3
25,1
25,3
25,7
26,6
27,1
27,6
28,4
28,4
28,7
29,0
29,8
30,1
30,3
31,1
31,4
31,7
32,0
32,4
33,0
33,2
33,4
33,8
34,5
34,8
36

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Reflection [Peakmaxima 2Theta]
Hydrat
35,1
35,9
37,0
37,1
37,4
37,5
38,0
38,3
38,5
38,8
39,1
39,3
Figure 1 shows the X-Ray powder diffractogram (at 25 C and with Cu-K alpha 1
as radiation source) of
the compound of formula (I) in the hydrate form.
In accordance with a fourth aspect, the present invention relates to use of a
compound selected
from:
I
F3C1\1
HH
\
,N
N
H
(11a) , and
37

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
I
N/rC)
F3C
H
\
0y1iL ,N N
H
0
(Vila) ,
for preparing a compound of formula (I):
, I F3Ci\rr n \-1
HO
H 0_, N j ( OH
N
(I) ,
or crystalline needles of the compound of formula (I) as described supra,
by the method as described supra. In accordance with a fifth aspect, the
present invention relates to
use of a compound of structure:
c3iF1
Ts
(VI)
,
for preparing a compound of formula (I):
, I F3Ci\rr n \-1
HO H 0, N j ( OH
N
(I) ,
38

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
or crystalline needles of the compound of formula (I) as described supra.
39

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Methods
HPLC:
Method A
HPLC instruments used:
a) Agilent Technologies 1260 Infinity
b) Agilent 1100 Series
Zorbax SB-AQ, 50*4.6 mm, 1.5 um
Buffer: Ammonium dihydrogenphosphate pH: 2.4
Acetonitrile
0 min. 5% buffer
8.3 min 80% buffer
11 min. 80% buffer
210 nm / 4 nm
1.2 ml! min.
Method B
HPLC Instrument used: Agilent Technologies 1260 Infinity
Al: Acetonitrile
B1: 2.72 g KH2PO4+ 2.32 g H3PO4+ 2 L H20
Agilent Poroshell 120 EC-C18 3*50mm 2.7u
Low Pressure Limit: 0.00 bar
High Pressure Limit: 400.00 bar
Flow: 1.000 mL/min
Maximum Flow Gradient: 1.000 mL/min2
Stop time: 8.00 min
Post time: 5.00 min
Starting conditions: A: 5% B: 95%

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Timetable
Time A B Flow Pressure
min % mL/min bar
8.00 80.0 20.0 1.000 400.00
Injection Volume: 5.00 uL
Temperature (Column): 45.00 C
Signal Wavelength: 210 nm
Method C
HPLC instrument used:
Agilent Technologies, HPLC 1290 Infinity (with DAD)
Apparatus 1. Ultra-High performance liquid
chromatograph
thermostatically controlled column oven, UV-
detector and data evaluation system
2. Stainless steel column
Length: 5 cm
Internal diameter: 2.1 mm
Filling: Acquity UPLC C18
BEH,
1.7 pm
Reagents 1. Acetonitrile, for the HPLC
2. Water, analytical grade
3. Phosphoric acid 85%, analytical grade
41

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Test solution Dissolve the sample in acetonitrile in a
concentration
of 0.25 mg/mL.
(e. g. dissolve approx. 25 mg sample, accurately
weighed in acetonitrile 100 mL.)
Calibration solution Dissolve the reference standard* in
acetonitrile in a
concentration of 0.25 mg/mL.
(e. g. dissolve approx. 25 mg reference standard,
accurately weighed, in acetonitrile 100 mL.)
* reference standard means the compound, which has
to be analyzed, as highly pure compound, i.e. >97
area% HPLC
Control solution Prepare a control solution that is identical
with the
calibration solution. Additionally, the control solution
contains small amounts of the organic impurities.
Detection sensitivity solution Prepare a solution containing the component
Solbrol P
(CAS-no.: 94-13-3; propyl 4-hydroxybenzoate) (RT
approx. 2.75 min) diluted to a concentration of 0.35
ug/m L.
HPLC conditions The specified conditions are guide values. To
achieve
optimal separations they should, if necessary, be
adapted to the technical possibilities of the
chromatograph and the properties of the respective
column.
Eluent A. 0.1 % Phosphoric acid 85% in water
42

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
B. Acetonitrile
Flow rate 1.0 mL/min
Temperature of the column oven 40 C
Temperature of the sample room temperature
chamber
Detection Measuring wavelength: 220 nm
Bandwidth: 6 nm
Injection volume 2.0 uL
Draw speed 200 uL/min
Needle wash Solvent for flush port: acetonitrile
Data rate 10 Hz
Cell dimension 10 mm
Equilibration time 10 min (at starting conditions)
43

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Time [min] %A %B
Gradient
0 95 5
2 70 30
6 60 40
8 20 80
12 20 80
Runtime of the chromatogram 12 min
Calculation of assay (content) The
assay is calculated using linear regression and
taking into account the sample weight and assay and
weight of the reference standard, with a validated
chromatographic data system
(e. g. Empower).
Method D
HPLC Instrument used: Agilent Technologies 1260 Infinity
Al: Acetonitrile
B1: 1.36 KH2PO4+ 1.74 K2HPO4+ 2 L H20
Eclipse XDB-C18 3*150mm 3,5u
Low Pressure Limit: 0.00 bar
High Pressure Limit: 400.00 bar
Flow: 0.500 mL/min
Stop time: 35.00 min
Post time: 10.00 min
Starting conditions: A: 95% B: 5%
Timetable
Time A B Flow Pressure
min % % mL/min bar
30.00 20.0 80.0 0.500 400.00
35.00 20.0 80.0 0.500 400.00
44

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Injection Volume: 3.004
Temperature (Column): 35.00 C
Signal Wavelength: 220 nm
GC-HS
Residual solvent analysis via headspace gas chromatography (GC-HS)
Agilent 6890 gas chromatograph with split-injection and FID (column: Restek
Rxi Sil MS; length:
20 m; internal diameter: 0.18 mm; df = 1 um). Injector temp 160 C, flow 1.2
ml/min (H2) Split Ratio
18, oven Temp 40 C (4.5min) ¨ 14 C/min ¨ 70 C ¨ 90 C/min ¨ 220 C (1.69 min).
Detector: temp
300 C, 400 ml/min (synth air), 40 ml/min (H2), 30 ml/min (N2), rate 20 Hz.
Perkin Elmer Turbomatrix 40 headspace sampler: oven 80 C, needle 150 C,
transfer line 160 C,
system pressure 140 kPa, equilibration time 32 min, pressurization 4.0 min,
injection time 0.04
min (Sampler) 0.05 min (GC).
Sample concentration: 20 mg substance in 2 ml DMF
Particle Size Analysis
The particle size analysis is done according to European Pharmacopeia 2.9.31
The equipment was developed and manufactured by Sympatec GmbH.
The components are as follows:
= RODOS dry dispersing system with turntable and spinning brush
= HELOS laser optical bench system with detector and data acquisition units
= HELOS software for system control, data transformation and report
generation
N-[2-(3-hydroxy-3-methyl butyl)-6-(2-hydroxypropan-2-y1)-2H-indazol-5-y1]-6-
(trifluoromethyl)-
pyridine-2-carboxamide (I) in its crystalline form A is applied on the
turntable. The particles are
brushed into a stream of pressurized air and dispersed. When passing the laser
beam the aerosol
generates a diffraction pattern, which is detected and analyzed according to
the Fraunhofer
model (European Pharmacopoeia 8.0, 2.9.31. Particle Size Analysis by Laser
Light Diffraction,
01/2010:20931, page 333 - 336). The results are formatted after user selection
for display and
printout of tables and graphics. The data are reported in um and volume
percent.

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
System settings
dispersion medium: dry air
air pressure: 4.0 bar
focus: 100 mm
airflow: 2.6 m3/ h
optical density: 3 - 12 %
detection time: min. (not less than) 1 s
rotation: 18 %
sample amount: approx. 200 mg
For routine purposes the mean of three measurements is reported.
HPLC Trace Analysis (ppm)
Instrument used: ultra-high performance liquid chromatograph (Agilent 1290)
equipped with a
thermostatically controlled column oven, mass spectrometer (Agilent 6420
Triple Quad-MS), UV-
detector and data evaluation system
Column Zorbax Eclipse Plus C8
Length: 50 mm
Internal diameter: 2.1 mm
Particle size: 1.8 um
Temperature: 40 C
Mobile Phase Eluent A 0.1% aq. formic acid
(compressibility: 45*10-6/bar)
Eluent B Acetonitrile contains 0.1% formic
acid
(compressibility: 120*10-6/bar)
46

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Flow 0.8 mL/min
Test solution Dissolve the sample in methanol in a concentration
of 10.0
mg/mL.
(e. g. dissolve approx. 20 mg sample, accurately weighed in
methanol 2 mL.)
Calibration solutions Dissolve a characterized standard of (VI) in
methanol in
concentrations of 0.2, 0.3, 0.4, 0.5, 0.6 and 0.75 g/mL.
Temperature of the column 40 C
oven
Temperature of the 10 C
autosampler
Detection (not used for Measuring wavelength: 220 nm
quantification)
Bandwidth: 6 nm
Injection volume 1.5 uL
Data rate 2.5 Hz
Detector cell 10 mm
Equilibration time 5 min (at starting conditions)
47

CA 03022327 2018-10-26
WO 2017/186693 PCT/EP2017/059748
Time [min] %A %B
Gradient
0.0 80 20
7.5 60 40
10.0 20 80
12.0 20 80
Runtime of the 12 min
chromatogram
MSD parameters (used for The conditions described here are applicable with
Agilent
quantification) 6420 Triple Quad-MS
Ion source Electrospray ionisation (ESI)
Time filtering Peakwidth 0.07 mm
Multiple reaction monitoring Precursor ion 281.1, product ion 194.9
used for quantification
Fragmentor 85 V
Collison energy 5 V
Source parameters
Gas temperature 350 C
Drying gas 13 L/min
Neb. Press. 50 psi
VCap 3000 V
Recovery For determing the recovery (W) a sample is spiked
with a
calibration solution of (VI) and then subjected to
measurement
Equation for calculating the GAP - Gp
W = ______________________________________________ = 100%
percentage of recovery GA
W = Recovery [h]
GAP = Content of (VI) in spiked sample
Gp = Content of (VI) in sample
GA = Spiked amount of (VI)
48

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Calculation of content of (VI) G = (P)1)i b WP,soll
(P)i
in sample a (WP)
(G)1 = content of (VI) in ith sample
(Pp)i = peak area of (VI) in ith sample
(VI7 p) = weight of it sample
Wh P,soll = target weight of it sample
a = slope of calibration curve
b = axis intercept of calibration curve
X-ray crystallography: measurement conditions:
Anode material Cu
K-Alpha1 [A] 1,54060
Generator settings 40 mA, 40 kV
Primary beam monochromator focussing X-ray mirror
Rotated sample Yes
Scan axis Gonio
Start Position [ 2Th.] 2.0066
End Position [ 2Th.] 37.9906
49

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Working Examples
The following examples illustrate the present invention.
Example #1
Methyl 5-(1[6-(trifluoromethyl)pyridine-2-yl]carbonyl}amino)-1H-indazole-6-
carboxylate (Vila)
Variant #1
30 g methyl 5-amino-1H-indazole-6-carboxylate (XII) along with 28.5 g 6-
(trifluoromethyl)pyridin-
2-carboxylic acid (XI) were suspended in 235 ml (210g) THE at 20- 25 C. 40m1
(30.4g) N,N-
diisopropylethylamine were added. The mixture, a yellow solution, was then
cooled to 0 C. To
this mixture, 187 ml (199.7 g) of a 50 wt% solution of propylphosphonic
anhydride (T3P) in ethyl
acetate were added over 45 min at 0 C. The dropping funnel was rinsed with 17
ml (15 g) THE.
After complete addition, the reaction mixture was stirred for 2 h at 0 C. The
solution had turned
red. The cold reaction mixture was then dropped over 45 min to 1.2 L water
kept at 1.5 C. The
dropping funnel was rinsed with 17 ml (15 g) THE. The pH of the mixture was
determined to be at
pH 1.6 (pH 1-2). The pH of the mixture was then adjusted to 7.5 via addition
of 45 ml (40 g) of a
28-30 wt% ammonium hydroxide solution at 1.5 C. Stirring was continued for 1
h at 1.5 C. The
resulting suspension was then warmed to ambient temperature (20 - 25 C)
within 1 h and stirring
was continued for 15 min. The precipitate was filtered off and washed with 100
ml water and
subsequently with 2 x 76 ml (60 g) ethanol. The product was dried in a drying
oven under vacuum
(160 mbar) and N2-flux at 45 C for 22 h.
Yield: 52.8 g (92.4 %, purity: 99.3 area% HPLC)
HPLC (Method B): Rt = 5.6 min.
MS (ESI pos): m/z = 365 (M+H)+
1-I-1 NMR (500 MHz, DMSO-d6): 6 [ppm]: 3.98 (s, 3 H), 8.21 (d, 1H), 8.25 (s,
1H), 8.31 (s, 1H), 8.39 (t,
1H), 8.48 (d, 1H), 9.16 (s, 1H), 12.57 (s, 1H), 13.45 (br s, 1H).
1-I-1 NMR (300 MHz, DMSO-d6): CI [ppm] = 3.97 (s, 3 H), 8.13 -8.27 (m, 2 H),
8.30 (s, 1 H), 8.33 - 8.45
(m, 1 H), 8.45 -8.51 (m, 1 H), 9.15 (s, 1 H), 12.57 (s, 1 H), 13.44 (br s, 1
H).
This procedure was carried out at a technical scale using 2.5 kg of (XII). Two
reactions were
performed at this scale. Each reaction was split into 4 batches for work-up
and isolation:
50

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Table 2: Batches and yields after manufacturing of (Vila) from (XII)
Reaction # Batch # Yield
1.007 kg
1
84.6 %
1.111 kg
2
93.3 %
1 (2.5 kg scale)
1.051 kg
3
88.2 %
1.055 kg
4
88.6 %
1.041 kg
87.4 %
1.123 kg
6
94.3 %
2 (2.5 kg scale)
1.056 kg
7
88.7 %
1.048 kg
8
88.0 %
Variant #2
5 2000 g (10,46 mol) methyl 5-amino-1H-indazole-6-carboxylate (XII), 1899 g
(9.94 mol)
6-(trifluoromethyl)pyridinee-2-carboxylic acid (XI) und 2028 g (15.69 mol) N,N-

diisopropylethylamine were mixed in 14.2 kg THE. At 0 - 5 C, 13.3 kg of a
solution of T3P in ethyl
acetate (50 wt%) was added dropwise within 30 min. Stirring was continued for
2 h at the same
temperature.
Work-Up:
The reaction mixture was warmed to ambient temperature (20 C). 3000 g of
water were added
while the temperature was kept at 20 - 25 C. Stirring was continued for 10
min. The pH was
adjusted to ca. 7.4 (7-8) using 4 N aq. sodium carbonate solution. Stirring
was continued for 10
min. If necessary the pH was again adjusted to 7.4 using 4 N aq. sodium
carbonate solution.
The solvents (THE/ethyl acetate) were evaporated under reduced pressure (¨ 200
mbar, 45-50 C
internal temperature) until the limit of stirring was reached. A mixture of
4.7 kg ethanol and
51

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
14.0 kg water was added and the pH was again adjusted to pH 7.4 (7-8) using 4
N aq. sodium
carbonate solution.
The mixture was stirred for 1 h at 50 C, subsequently cooled to 20 - 25 C.
Stirring was continued
for 10 min at the same temperature. The precipitated crystals were filtered,
washed with a
mixture of ethanol and water (1.3 kg ethanol with 4 kg water) and dried under
vacuum in a drying
oven (45 C, N2 flux, at least 12 h).
According to the above described procedure, four batches using 2 kg of
starting material (methyl
5-amino-1H-indazole-6-carboxylate) were produced in the technical laboratory:
Yields:
Batch #1: 3476 g (95 %)
Batch #2: 3449 g (95 %)
Batch #3: 3476 g (95%)
Batch #4: 3494 g (96%)
The purities of all batches were determined to be > 98 area% (H PLC).
HPLC (Method A): Rt = 6.5 min.
MS (ESI pos): rniz = 365 (M+H)+
1-I-1 NMR (500 MHz, DMSO-d6): 6 [ppm]: 3.98 (s, 3 H), 8.21 (d, 1H), 8.25 (s,
1H), 8.31 (s, 1H), 8.39 (t,
1H), 8.48 (d, 1H), 9.16 (s, 1H), 12.57 (s, 1H), 13.45 (br s, 1H).
1-I-1 NMR (300 MHz, DMSO-d6): 6 [ppm] = 3.97 (s, 3 H), 8.13 -8.27 (m, 2 H),
8.30 (s, 1 H), 8.33 - 8.45
(m, 1 H), 8.45 -8.51 (m, 1 H), 9.15 (s, 1 H), 12.57 (s, 1 H), 13.44 (br s, 1
H).
Example #2
N-[6-(2-hydroxypropan-2-y1)-1H-indazol-5-y1]-6-(trifluoromethyl)pyridine-2-
carboxamide (11a)
In the following section, different variants of the reaction procedure and
work-up are described.
These procedures are oriented at the given conditions in the respective
technical plants. The
following experiments were performed at the exclusion of water and air using
inert gas (N2 or Ar).
52

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Variant #1
50 g (137.26 mmol) of methyl 5-({[6-(trifluoromethyppyridin-2-
yl]carbonyllamino)-1H-indazole-6-
carboxylate (Vila) were dissolved in 800 ml THE. Under normal pressure (1 atm)
ca. 300 ml THE
were distilled off at 70 C. The solution was then cooled to 0 - 3 C.
The solution was kept at this temperature and added dropwise within 120 min to
a cooled
mixture of 457.5 ml (1372.55 mmol) methylmagnesium chloride 3 M in THE and
29.1 g lithium
chloride (686.27 mmol) at 0 - 3 C. After the addition was complete, a sample
was taken out of the
mixture and subjected to HPLC analysis showing that conversion was complete.
The mixture was
poured carefully over 25 min at 0 - 3 C into 500 ml half-sat. aqu. sodium
chloride solution
(attention: exothermic! During the first 50 ml a strong rise in temperature to
29 C was
observed!). A suspension was received which dissolved when 358 ml 20 wt% aq.
citric acid were
added (pH dropped from 8.08 to 4.28). Stirring was continued for 10 min at 20 -
25 C. 500 ml of
ethyl acetate were added and stirring was continued for 10 min. The phases
were separated. The
mulm was added to the organic phase. 5 g of activated charcoal were added to
the organic phase.
The mixture was heated to 78 C (internal temperature), stirred for 30 min at
that temperature
and subsequently cooled to 50 C (internal temperature). The warm solution was
filtered over
celite and washed twice with 125 ml ethyl acetate. The mixture was
concentrated to ca. 150 ml at
ambient pressure (1 atm) and 110 C. 350 ml of toluene were added and 200 ml
were distilled off
at ambient pressure (1 atm) and 110 C. The product precipitated. At 60 C
internal temperature,
200 ml n-heptane were added over 45 min. The mixture was cooled to 0 - 3 C
and stirred for 2 h
at this temperature. The product was filtered and washed twice with a mixture
of 50 ml
toluene/n-heptane (1:1). The precipitated product was dried in a drying oven
at 40 C and
20 mbar for >48 h.
Yield: 39,42 g (78,83 %, purity 97,84 area% HPLC)
HPLC (Method A): Rt = 5.8 min.
MS (ESIpos): rn/z = 365 (M+H)+
1-1-1-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.63 (s, 6H), 5.99 (s, 1H), 7.50 (s,
1H), 8.06 (s, 1H),
8.17 (d, 1H), 8.37 (t, 1H), 8.46 (d, 1H), 8.78 (s, 1H), 12.33 (s, 1H), 12.97
(br s, 1H).
13 batches were produced following the procedure of variant #1. The table
below summarizes the
respective yields. The reactions were performed at 1 kg scale with regard to
the use of methyl 5-
({[6-(trifluoromethyl)pyridine-2-yl]carbonyllamino)-1H-indazole-6-carboxylate
(Vila) as starting
material. In most cases, two batches were united after treatment with
activated charcoal:
53

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Table 3: Batches and yields after manufacturing of (11a) from (Vila)
Batch # Yield [kg]
Fol
1 1.597 kg
2 79.9%
3 1.88 kg
4 94%
1.816 kg
6 90.8%
7 1.66 kg
8 83%
9 1.752 kg
87.6%
11 1.854 kg
12 92.7%
0.919 kg
13*
96.4 %
*) single batch
5 Variant #2
30 g (82,353 mmol) methyl 5-({[6-(trifluoromethyppyridine-2-yl]carbonyllamino)-
1H-indazole-6-
carboxylate (Vila) were dissolved in 480 ml THE. Under normal pressure (1 atm)
ca. 180 ml THE
were distilled off at 70 C. The mixture (slight suspension) was then cooled
to 0 - 3 C.
The solution was kept at this temperature and added dropwise within 120 min to
a cooled
10 mixture of 274.5 ml (823.528 mmol) methylmagnesium chloride 3 M in THE
and 17.5 g lithium
chloride (411.764 mmol) at 0 - 3 C. 15 min after the addition was complete, a
sample was taken
out of the mixture and subjected to HPLC analysis (method A) showing that (VI)
was completely
converted. The mixture was poured carefully over 15 min at 0 - 3 C into 300
ml of water
(attention: exothermic! During the first 50 ml a strong rise in temperature
was observed!). 310 ml
20 wt% aq. citric acid were added (pH dropped to 4.05). Stirring was continued
for 60 min at
54

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
20 to 25 C. 300 ml of ethyl acetate were added and stirring was continued for
30 min. The
phases were separated. The mulm was added to the organic phase. The organic
phase was
washed twice with 450 ml of water. The organic phase was concentrated to 350
ml at 65 C
(internal temperature) and ambient pressure (1 atm). 250 ml ethyl acetate were
added. 6 g of
activated charcoal were added to the organic phase. The mixture was heated to
65 C (internal
temperature), stirred for 120 min at that temperature and subsequently cooled
to 50 C (internal
temperature). The warm solution was filtered over celite and washed twice with
125 ml ethyl
acetate. The mixture was concentrated to ca. 150 ml at ambient pressure (1
atm) and 110 C.
300 ml of toluene were added and 200 ml were distilled off at ambient pressure
(1 atm) and
110 C. The product precipitated. At 60 C internal temperature, 200 ml n-
heptane were added
over 45 min. The mixture was cooled to 0 - 3 C and stirred for 2 h at this
temperature. The
product was filtered and washed twice with a mixture of 50 ml toluene/n-
heptane (1:1). The
precipitated product was dried in a drying oven at 40 C and 20 mbar for >48
h.
Yield: 24,0 g (80%, purity: 95,8 area% H PLC)
HPLC (Method A): Rt = 5.8 min.
MS (ESI pos): rn/z = 365 (M+H)+
1-1-1-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.63 (s, 6H), 5.99 (s, 1H), 7.50 (s,
1H), 8.06 (s, 1H),
8.17 (d, 1H), 8.37 (t, 1H), 8.46 (d, 1H), 8.78 (s, 1H), 12.33 (s, 1H), 12.97
(br s, 1H).
Variant #3
g (82.353 mmol) methyl 5-({[6-(trifluoromethyppyridine-2-yl]carbonyllamino)-1H-
indazole-6-
carboxylate (Vila) were dissolved in 600 ml THE. Under normal pressure (1 atm)
ca. 150 ml THE
were distilled off at 70 C. The mixture (slight suspension) was then cooled
to 0 - 3 C.
The solution was kept at this temperature and added dropwise within 120 min to
a cooled
25 mixture of 274.5 ml (823.528 mmol) methylmagnesium chloride 3 M in THE
and 17.5 g
(411.76 mmol) lithium chloride at 0 - 3 C. The dropping funnel was rinsed
twice with 10 ml THE.
15 min after the addition was complete, a sample was taken out of the mixture
and subjected to
HPLC analysis showing that (Vila) was completely converted. The mixture was
poured carefully
over 10 min at 0 - 3 C into 300 ml of water (attention: exothermic! During
the first 50 ml a strong
30 rise in temperature to 25 C was observed!). 250 ml 20 wt% aq. citric
acid were added (pH

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
dropped from 8 to 4). Stirring was continued for 30 min at 20 - 25 C. 300 ml
of ethyl acetate were
added and stirring was continued for 10 min. The phases were separated. The
mulm was added to
the organic phase. The organic phase was washed twice with 200 ml of 1wt%
sodium chloride aq.
solution. The phases were separated. The organic phase was concentrated to 250
ml at 65 C
(internal temperature) and ambient pressure (1 atm). 150 ml ethyl acetate and
6 g of activated
charcoal were added to the organic phase. The mixture was heated to 65 C
(internal
temperature), stirred for 120 min at that temperature and subsequently cooled
to 50 C (internal
temperature). The warm solution was filtered over celite and washed twice with
50 ml ethyl
acetate. The mixture was concentrated to ca. 100 ml at ambient pressure (1
atm) and 110 C.
300 ml of isopropanol were added. 300 ml were distilled off at ambient
pressure (1 atm) and
110 C. 300 ml isopropanol were added again and distilled off (ca. 355 ml) at
110 C. The resulting
suspension was cooled to 20-25 C. 45 ml water were added over 45 min. The
mixture was stirred
for 1 h. The precipitated product was filtered and washed with 50 ml of a
water/isopropanol (1:1)
mixture. The precipitated product was dried in a drying oven at 50 C and 20
mbar for >48 h.
Yield: 24,9 g (83 %, purity: 97,84 area% HPLC)
HPLC (Method A): Rt = 5.8 min.
MS (ESI pos): rn/z = 365 (M+H)+
11-1-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.63 (s, 6H), 5.99 (s, 1H), 7.50 (s, 1H),
8.06 (s, 1H),
8.17 (d, 1H), 8.37 (t, 1H), 8.46 (d, 1H), 8.78 (s, 1H), 12.33 (s, 1H), 12.97
(br s, 1H).
Variant #4
This variant was used for the production of technical batches at kg scale (>10
kg).
60 g (164.706 mmol) methyl 5-({[6-(trifluoromethyppyridine-2-
yl]carbonyllamino)-1H-indazole-
6-carboxylate (Vila) were dissolved in 1500 ml THE. Under normal pressure (1
atm) ca. 600 ml THE
were distilled off at 70 C. The mixture (yellow solution) was then cooled to
0 - 3 C.
The solution was kept at this temperature and added dropwise within 120 min to
a cooled
mixture of 550 ml (1647.06 mmol) methylmagnesium chloride 3 M in THE and 35 g
(823.53 mmol)
lithium chloride at 0 - 3 C. 15 min after the addition was complete, a sample
was taken out of the
mixture and subjected to HPLC analysis showing that the conversion of (Vila)
was complete. The
56

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
mixture was poured carefully over 15 min at 0 - 3 C into 600 ml of water
(attention: exothermic!
During the first 50 ml a strong rise in temperature was observed!). 600 ml 20
wt% aq. citric acid
were added (pH dropped to 4). Stirring was continued for 30 min at 20 - 25 C.
The phases were
separated. The organic phase was washed twice with 400 ml of 1 wt% sodium
chloride aq.
solution. The mulm was added to the organic phase. The phases were separated.
The organic
phase was concentrated to 700 ml at 65 C (internal temperature) and ambient
pressure (1 atm).
500 ml ethyl acetate and 12 g of activated charcoal were added to the organic
phase. The mixture
was heated to 65 C (internal temperature), stirred for 120 min at that
temperature and
subsequently cooled to 50 C (internal temperature). The warm solution was
filtered over celite
and washed twice with 200 ml ethyl acetate. Concentration was continued under
reduced
pressure (200 mbar). A solvent swap to touluene was performed (remaining
volume ca. 850 mL).
The resulting suspension was cooled to 0 - 3 C. The precipitated product was
filtered and washed
with 50 ml of toluene. The precipitated product was dried in a drying oven at
50 C and 20 mbar
for >48 h.
Yield: 51.2 g (85.3 %, purity: 96,.51 area% HPLC)
HPLC (Method A): Rt = 5.8 min.
MS (ESI pos): rn/z = 365 (M+H)+
1-1-1-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.63 (s, 6H), 5.99 (s, 1H), 7.50 (s,
1H), 8.06 (s, 1H),
8.17 (d, 1H), 8.37 (t, 1H), 8.46 (d, 1H), 8.78 (s, 1H), 12.33 (s, 1H), 12.97
(br s, 1H).
Variant #5
Purification via stirring in isopropanol/water
Depending on the purity of the crude product, an additional purification step
via stirring in
mixtures of isopropanol and water, preferably 1:1, can be performed. Depending
on the purity of
the crude product, stirring is performed in a range of 2 - 10 volumes with
regard to the crude
starting material. The following example describes stirring in 3 volumes
isopropanol/water:
7.5 g N46-(2-hydroxypropan-2-y1)-1H-indazol-5-y1]-6-(trifluoromethyppyridine-2-
carboxamide (11a)
with a purity of 95 area% (H PLC) were stirred in 22.5 ml of a 1:1 (vol)
mixture of water and
isopropanol for 2 h at 20 C. The suspension was then filtered and the product
washed with 4 ml
57

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
of the same solvent mixture. The product was dried in drying oven at 50 C
under vacuum
(<100 mbar).
Yield: 6.8 g (90.7 %, purity > 98 area% HPLC)
HPLC (Method A): Rt = 5.8 min.
MS (ESIpos): rniz = 365 (M+H)+
1-1-1-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.63 (s, 6H), 5.99 (s, 1H), 7.50 (s,
1H), 8.06 (s, 1H),
8.17 (d, 1H), 8.37 (t, 1H), 8.46 (d, 1H), 8.78 (s, 1H), 12.33 (s, 1H), 12.97
(br s, 1H).
Example #3
3-Hydroxy-3-methylbuty1-4-methylbenzenesulfonate (VI)
Variant #1
This variant was used for the production of technical batches at kg scale.
To a solution of 100 g 3-methylbutane-1,3-diol (IX) in 200 ml (264 g)
dichloromethane were added
147 ml (107 g) triethylamine along with 6.0 g 4-dimethylaminopyridine (DMAP).
The reaction
mixture was then cooled to 0 C (0 5 C).
In parallel, 192 g of 4-toluenesulfonyl chloride (X) were dissolved in 400 ml
(528 g)
dichloromethane. The resulting slightly cloudy solution was then dropped over
1.5 h to the
reaction mixture at 0 - 5 C. When the temperature of the reaction reached 5
C, the addition was
paused and continued when the internal temperature had dropped to 0 C. After
complete
addition, the reaction mixture was warmed to ambient temperature (20 - 25 C)
over 1 h. The
reaction mixture was then continuously stirred at ambient temperature for 12 -
18 h (preferably
15 h).
Subsequently, 500 ml of water were added to the reaction mixture. The mixture
was stirred for
additional 2 h at 20 - 25 C. The phases were separated. The mulm was
collected in the aqueous
phase. 500 ml of water were added to the organic phase and the pH was adjusted
to 1.9 using
5 ml 2 N aq. HCI. After phases were separated, 500 ml 1/2-saturated aq. NaCI-
solution was added
to the organic phase. The pH was adjusted to 7 using sat. aq. NaHCO3-solution.
The phases were
separated and the organic phase was concentrated via rotary evaporation in
vacuo (down to
14 mbar) at 40 C. The product was obtained as viscous yellow oil.
58

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Yield: 222.3 g (89.6 %, purity: 91.9 area% HPLC)
HPLC (Method A): Rt = 5.3 min.
MS (ESI pos): rniz = 241 [M-OH]
1-1-1-NMR (500MHz, DMSO-d6): 6 [ppm]= 1.12 (s, 6H), 1.78 (t, 2H), 2.50 (s,
3H), 4.20 (t, 2H),
4.47 (br s, 1H), 7.56 (d, 2H), 7.87 (d, 2H).
This procedure was carried out at a technical scale using 1.5 kg of (IX). Nine
batches were
produced. An overview is given in the table below.
Table 4: Batches and yields after manufacturing of (VI) from (IX)
Batch # (1.5 kg scale) Yield
3.477 kg
1
3.521 kg
2
94.6 %
3.458 kg
3
92.9 %
3.487 kg
4
93.7 %
3.499 kg
5
94.0%
3.490 kg
6
93.8 %
3.492 kg
7
93.8 %
3.624 kg
8
97.4 %
3.467 kg
9
93.2%
59

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Variant #2
400 g 3-methylbutane-1,3-diol were emulsified in 607 ml (528 g) toluene at
ambient temperature
(20 - 25 C). The emulsion was cooled to 0 C. 589 ml (427.5 g) of
triethylamine were added over
15 min (slightly exothermic). 23.5 g 4 dimethylaminopyridine (DMAP) were
added. Within 10 min
the reaction mixture had turned into a solution.
In parallel, 768.8 g of 4-toluenesulfonyl chloride were dissolved in 1214 ml
(1056 g) toluene
(endothermic!). The resulting slightly cloudy solution was filtered and the
filtrate was dropped
within 2 h to the reaction mixture at 0 C. After complete addition, stirring
was continued at 0 C
for 12-18 h (preferably 15 h). A white precipitate had formed
(triethylammonium chloride). The
precipitate was filtered off and the resulting clear solution (2603 g) was
used as a 30-35 wt%
solution of 3-hydroxy-3-methylbuty1-4-methylbenzenesulfonate (VI) in the
alkylation of N46-(2-
hydroxypropan-2-y1)-1H-indazol-5-y1]-6-(trifluoromethyppyridine-2-carboxamide
(11a) in
transformations analogous to example#5 variant#2.
HPLC (Method B): Rt = 4.68 min.
Variant #3
This variant was used for the production of technical batches at kg scale.
1.57 kg 3-methylbutane-1,3-diol (IX) were emulsified in 4.0 kg toluene at
ambient temperature
(20 - 25 C). 2 kg of solvent were distilled off at ambient pressure (T 3.10
C). The emulsion was
cooled to 0 C (internal temperature). 1.63 kg of trimethylamine and 89 g 4-
dimethylaminopyridine (DMAP) were added along with 0.1 kg toluene and stirred
for 15 min.
(slightly exothermic).
In parallel, 2.65 kg of 4-toluenesulfonyl chloride were dissolved in 3.7 kg
toluene (endothermic!,
therefore warmed to ambient temperature). The resulting slightly cloudy
solution was filtered
and the filter was washed with 0.11 kg toluene. The resulting filtrate was
dropped within 5 h to
the reaction mixture at 0 C. After complete addition, stirring was continued
at 0 C for 12-18 h
(preferably 15 h). A white precipitate had formed (triethylammonium chloride).
The precipitate
was filtered off and the precipitate washed with 3x 1.88 kg toluene. The
resulting clear solution
(14.4 kg) was determined to have a content of 25.4 wt% of 3-hydroxyl-3-methyl
butyl-4-
methylbenzenesulfonate (VI) and was used without further work-up in the
alkylation reaction of

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
N46-(2-hydroxypropan-2-y1)-1H-indazol-5-y1]-6-(trifluoromethyppyridine-2-
carboxamide (11a). This
solution was used in the transformation depicted in example#5 variant#3.
HPLC (Method C): Rt = 2.68 min.
Example #4
2-(3-Hydroxy-3-methylbuty1)-5-(1[6-(trifluoromethyl)pyridin-2-
yl]carbonyl}amino)-2H-indazole-
6-carboxylate (V)
This variant was used for the production of technical batches at kg scale.
1200 g of methyl 5-({[6-(trifluoromethyppyridin-2-yl]carbonyllamino)-1H-
indazole-6-carboxylate
(Vila), 12.0 L N,N-diisopropylethylamine and 7.5 L toluene were mixed at
ambient temperature
(20 - 25 C). The resulting yellow suspension was heated to an internal
temperature of 111 C
(120 C jacket temperature). A solution of 4255 g 3-hydroxy-3-methylbuty1-4-
methylbenzene-
sulfonate (VI) in 4.25 L toluene was slowly dosed to the reaction mixture over
10 h via syringe
pump. After complete addition, the dropping funnel was rinsed with 0.25 L
toluene. The reaction
mixture was then cooled to an internal temperature of 104 C and was stirred
at that temperature
for 12 - 18 h (preferably 15 h). The reaction mixture was then cooled to 45 C
(jacket
temperature). The volume of the reaction mixture was reduced at 45 C to 53 C
(jacket
temperature) under vacuum (113 - 70 mbar) to a viscous, well stirable residue
(ca. 19.6 L distillate
removed). At an internal temperature of 28 - 33 C (careful: prevent
crystallization by fast
addition of ethyl acetate) 12 L ethyl acetate were added followed by 12 L
water. The mixture was
stirred for 5 min at an internal temperature of 22 C. The phases were
separated. The mulm was
added to the aqueous phase. The aqueous phase was extracted with 3.85 L ethyl
acetate. The
organic phases were combined and 12 L of water were added. The pH of the
mixture was adjusted
from 10 to 6.9 (6 - 7) using conc. acetic acid. The organic phase was
evaporated to dryness at
40 C under vacuum (down to 45 mbar). The residue was dissolved in 1 L
dichloromethane and
evaporated to dryness. This was repeated two more times. The resulting residue
(1.772 kg) was
dissolved in 26.58 L dichloromethane (15 L/kg). The resulting solution was
adjusted to a
concentration of 20 L/kg (3.6 wt%) and subsequently subjected to column
chromatography
(chromasil 13 m; gradient: ethyl acetate/ n-hexane 10:90 to 100:0). The
resulting pure product
was provided as a 10-15 wt% solution in THE for the following step.
Four reactions were run at 1.2 kg scale each. These have been comprised in one
batch for column
chromatography. Further three reactions were run at the same scale and also
comprised in one
61

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
batch for column chromatography. The following table shows the results with
respect to yield and
purity:
Table 5: Yields and purity (HPLC) after manufacturing of (V) from (Vila)
Batch # Reaction # Yield Purity
(HPLC)
(1.2 kg scale (Vila))
1
2 3.39 kg
1 99.8 area%
3 47%
4
5
2.40 kg
2 6 99.5 area%
45%
7
HPLC (Method B): Rt = 5.9 min.
MS (ESI pos): rniz = 451 (M+H)+
1-1-1-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.16 (s, 6H), 2.00 - 2.13 (m, 2H), 3.96
(s, 3H), 4.45 -4.64
(m, 3H), 8.20 (d, 1H), 8.34 - 8.42 (m, 1H), 8.42 - 8.49 (m, 2H), 8.55 (s, 1H),
9.05 (s, 1H), 12.52 (s,
1H).
Alternatively, crystallization can be performed in order to obtain the
purified product as a neat
solid:
300 g of a 15 wt% solution of 2-(3-hydroxy-3-methylbutyl)-5-({[6-
(trifluoromethyppyridine-2-
yl]carbonyllamino)-2H-indazole-6-carboxylate (V) in THE was concentrated at 43
C jacket
temperature under vacuum (300 - 320 mbar). Distillation was continued until
the limit of
stirability was reached (199.6 g residue). At ambient pressure and a jacket
temperature of 43 C
255 g of n-heptane were added over 15 min to the residue. Stirring was
continued for 1 h before
the mixture was cooled to 20 C within 1 h. The mixture was stirred at that
temperature for 12 -
18 h (preferably 15 h). The product was filtered, washed twice with 25 g n-
heptane and dried in a
drying oven at 40 C under vacuum (<200 mbar).
62

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
Example #5
N12-(3-hydroxy-3-methylbuty1)-6-(2-hydroxypropan-2-y1)-2H-indazol-5-y1]-6-
(trifluoromethyl)-
pyridine-2-carboxamide (I)
Variant #1
The following experiment was performed at the exclusion of water and air using
inert gas (N2 or
Ar, preferably Ar).
4.0 kg anhydrous THE were placed in a reaction vessel under inert atmosphere
and cooled to
-15 C (internal temperature). 4.61 kg 3 M methylmagnesium chloride solution
in THE were
added. The dropping funnel was rinsed with 0.433 kg THE.
In parallel, 9.901 kg of a 10.1 wt% solution of methyl 2-(3-hydroxy-3-
methylbutyI)-5-({[6-
(trifluoromethyl)pyridine-2-yl]carbonyllamino)-2H-indazole-6-carboxylate (V)
was concentrated at
40 C under vacuum. App. 5 kg were distilled off and 2.087 kg residue
remained. To the residue
4.279 kg THE were added resulting in a 15 wt% solution of (V) in THE.
The 15 wt% solution of methyl 2-(3-hydroxy-3-methyl butyl)-5-({[6-
(trifluoromethyl)pyridin-2-
yl]carbonyllamino)-2H-indazole-6-carboxylate (V) in THE was slowly dosed over
at least 1 h 45 min
to the Grignard solution at -15 C. The container and pump were rinsed with
0.3 kg THE. Stirring
was continued for 30 - 40 min at the same temperature. Meanwhile, a 15 wt% aq.
solution of
citric acid (2.8 kg citric acid monohydrate + 14.267 kg water) was placed in a
reaction vessel and
cooled to 0 C (internal temperature). The cold reaction mixture (0¨ 10 C)
was dosed within 30
min to the aqueous citric acid solution. It was rinsed with 1 kg THE. The
quenched reaction
mixture was then allowed to warm to ambient temperature (20 - 25 C) over a
period of 40 min.
The phases were separated. The aqueous phase was extracted with 10 L ethyl
acetate. The
organic phases were combined and washed with 6.66 L water (phases were stirred
for 15 min).
The combined organic phases were concentrated until the limit of stirability
was reached (45 C
jacket temperature, vacuum 150 mbar to 70 mbar; app. 3 -4 L residual volume).
6 kg of ethanol
were added to the residue. The solution was concentrated under vacuum (45 to
max. 60 C jacket
temperature; 8.5 L distillate) and again 6 kg of ethanol were added. The
solution was again
concentrated under vacuum (distillate: 7.95 L). Then, 6 kg of ethanol were
added to the residue.
Crude crystallization:
The resulting solution was heated to an internal temperature of 31 - 32 C. 18
L water were added
within 1 h resulting in a yellowish suspension. The mixture was cooled to 20
C within 1 h and
stirred for 20 min. The precipitate was filtered and washed twice with a
mixture of 0.416 kg
63

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
ethanol + 1.25 kg water. The mother liquor was filtrated again and the
precipitate washed with a
mixture of 1.7 kg ethanol/water (1:3). The crude product was dried in a drying
oven at 40 C
under vacuum (<200 mbar) for 12 - 18 h (preferably 15 h).
Recrystallization (3 reactions (crude product batches) were combined in one
batch for
purification):
The combined crude products (2.855 kg) were suspended in 18.27 kg of a 9:1
mixture of
toluene/acetone. The mixture was then heated to 80 C internal temperature and
6.67 kg of a 9:1
mixture of toluene/acetone were added in portions of 1.1 L. Upon dissolution
of the product, the
mixture was cooled to 55 C. Then slowly cooled to 52 C and stirred for 1 h
at that temperature.
The product started to crystallize at 53 C. (Seeding with crystals is
optional). Stirring was
continued for 1 h at 52 C (internal temperature). The suspension was then
cooled within 2 h to
C. The suspension was stirred at 20 C for 12 - 18 h (preferably 15 h). The
product was filtered
and washed with 1.11 kg toluene/acetone 9:1 and subsequently with 1.11 kg
toluene. The
product was dried in a drying oven at 40 C under vacuum (<200 mbar) for 12 -
18 h (preferably
15 15 h).
In order to obtain a defined crystal habit the pure product is subjected to
crystallization with
ethanol and water (as described above, analogous to first crystallization from
ethanol/water).
Thus, needles of the product are obtained in high purity: 8.37 kg ethanol are
added to 2.32 kg of
the purified product. The mixture is warmed to 32 C. At that temperature 25.1
kg water are
20 added over a period of 1 h. The resulting suspension is cooled to 20 C
within 1 h and stirred for
20 min. The product is filtrated and washed with 7.43 kg of a mixture of
ethanol/water (1:3). The
precipitate is washed two more times with 7.43 kg of a mixture of
ethanol/water (1:3). The
product was dried in a drying oven at 50 C under vacuum (<200 mbar) for 12 -
18 h (preferably
15 h).
Table 6: Yields and purity (HPLC) after manufacturing of (I) from (V)
Batch # Reaction # Yield Purity (H
PLC)
(1.0 kg scale (V)) Content
1
2.314 kg 98.1 area%
1 2
77.1 % 97.92 %
3
4
2.164 kg 98.25 area%
2 5
72.1 % 97.96 %
6
64

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
HPLC (Method C): Rt = 3.50 min.
MS (ESI pos): rniz = 451 (M+H)+
1-1-1-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.15 (s, 6H), 1.62 (s, 6H), 1.99 - 2.08
(m, 2H), 4.45 ¨ 4.50
(m, 2H), 4.51 (s, 1H), 5.94 (s, 1H), 7.57 (s, 1H), 8.16 (d, 1H), 8.35 (s, 1H),
8.36 - 8.39 (m, 1H), 8.43 -
8.47 (m, 1H), 8.71 (s, 1H), 12.35 (s, 1H).
1H-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.15 (s, 6H), 1.63 (s, 6H), 2.00 - 2.09 (m,
2H), 4.43 - 4.55
(m, 3H), 5.94 (s, 1H), 7.57 (s, 1H), 8.16 (d, 1H), 8.34 - 8.39 (m, 2H), 8.45
(d, 1H), 8.72 (s, 1H), 12.36
(s, 1H).
Variant #2
An approximately 30 - 35 wt% solution of 3-hydroxy-3-methylbuty1-4-
methylbenzenesulfonate
(VI) in toluene was freshly prepared analogously to the procedure given in
example #3 variant #2.
100 g of N46-(2-hydroxypropan-2-y1)-1H-indazol-5-y1]-6-
(trifluoromethyppyridine-2-carboxamide
(11a) were suspended in 560.5 g toluene. The mixture was heated to 104 C (110
C) within 30 min.
Within 5 h, 212.8 g N,N-diisopropylethylamine and 1013 g of a 35 wt% solution
of (VI) in toluene
were dosed simultaneously to the reaction mixture within 5 h. Thereby, it is
important that an
excess of base is always present during the reaction. After complete addition,
the reaction
mixture was stirred at 104 C (110 C) overnight (18 h). The reaction mixture
(two phases had
formed) was then cooled to 45 C and concentrated under vacuum (down to app.
50 mbar) to a
viscous, stirrable residual volume of app. 750 ml (1189.9 g were distilled
off). The residue was
then cooled to 20 C and 920 g ethyl acetate were added followed by a mixture
of 110 g conc.
acetic acid and 840 g water. The mixture was stirred for 5 min at 20 C. The
phases were
separated. The aqueous phase was reextracted with first 840 g and then with
420 g ethyl acetate.
The organic phases were combined and 840 g water were added. Phases were
separated. The
phases were recombined and the mixture was heated to 50 C (internal
temperature) and stirred
for 1 hour at that temperature. Phases were separated and the organic phase
was concentrated
under vacuum at a temperature of 50 - 60 C to a residual volume of app.
213.4g.
840 g isopropanol were added to the residue. The solvents were evaporated to a
final residue of
app. 380.9 g in order to remove all remaining ethyl acetate. This procedure
can be repeated if
necessary. To the isopropanolic residue (380.9 g) were added 187.6 g of
isopropanol and 419 g of
isopropanol. This resulted in a 27.3 wt% solution of crude (1) in isopropanol
(purity: 78.4 area%
H PLC).

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
HPLC (Method C): Rt = 3.58 min.
316.9 g of this solution were used in the following precipitation procedure:
The solution was kept
at 25 C. Within 30 min 984.4 g of water were added. Seed crystals (1 %; 0.33
g) were added.
Stirring was continued for 30 min. Within 2 h 564 g of water were added. The
resulting suspension
was stirred for 1h and filtered. The precipitate was washed with a mixture of
15.4 g isopropanol
and 46.8 g water followed by 62.1 g water. The product is dried in a drying
oven at 50 C under
vacuum for 18 h.
Using this procedure, crude product was obtained in 81 % yield with a purity
of 89.2 area%
(84.4 wt%).
HPLC (Method C): Rt = 3.55 min.
Material obtained with the afore described work-up can be purified via
repetitive crystallization
from toluene/acetone 9:1 in the presence of activated charcoal similar to the
crystallization
described in the procedure for variant #1. A definite crystal form can be
obtained via
recrystallization with ethanol and water (see also procedure variant #1). An
example is given here:
23.0 g crude (I) (89 area% H PLC; 86 wt%; method D) were suspended in 70 g of
a toluene/acetone
mixture (9:1). The mixture is heated to 80-82 C internal temperature (slight
reflux observed). 87 g
of the toluene/acetone mixture (9:1) were added. A clear solution resulted.
4.6 g of activated
charcoal were added. Stirring was continued for 30 min at that temperature.
The hot solution was
filtrated over 2.5 g harbolite 900. The filter was rinsed with 9.5 g of the
toluene/acetone mixture
(9:1). Crystallization in the filtrate started at 60 C. The mixture was
stirred at 60-62 C internal
temperature for 1 h. The suspension was then cooled to 22 C within 2.5 h and
stirred for
app. 16 h (overnight). The purified product was filtrated and washed with 20 g
of the
toluene/acetone mixture (9:1) and dried in a drying oven under vacuum at 50 C
for 24 h.
Yield: 14.9 g (64.8%; purity: 96.2 area% HPLC; 94.1 wt%)
HPLC (Method C): Rt = 3.47 min.
14.9 g of purified product were obtained of which 13.6 g were again subjected
to recrystallization:
13.6 g purified (I) were suspended in 85.7 g of a toluene/acetone mixture
(9:1). The mixture is
heated to 80 to 82 C internal temperature. 32.7 g of the toluene/acetone
mixture (9:1) were
added. A clear solution resulted. 2.8 g of activated charcoal were added.
Stirring was continued
for 30 min at that temperature. The hot solution was filtrated over 2.5 g
harbolite 900. The filter
was rinsed with 10 g of the toluene/acetone mixture (9:1). Crystallization in
the filtrate started at
70 C. The mixture was stirred at 70 C internal temperature for 1 h. The
suspension was then
66

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
cooled to 22 C within 4 h and stirred for app. 18 h. The purified product was
filtrated and washed
with 10 g of the toluene/acetone mixture (9:1) and dried in a drying oven
under vacuum at 50 C
for 24 h.
Yield: 11.5 g (84.6%; purity: 97.7 area% HPLC; 91.5 wt%)
HPLC (Method C): Rt = 3.48 min.
11.5 g of a the purified product were obtained of which 9 g were subjected to
crystallization with
ethanol/water for obtaining the right crystal form and removing inclusions of
toluene (7.3 wt%):
To 9.0 g of purified (1) 32.4 g ethanol were added and the mixture was warmed
to 32 C (internal
temperature). 92.7 g water were added to the solution within 1 h. The
resulting suspension was
stirred for 30 min at that temperature. The suspension is cooled to 22 C
within 1 h. The
crystalline product was filtrated and washed with a mixture of 6.6 g water and
3.3 g ethanol and
dried in a drying oven under vacuum at 50 C for 24 h.
Yield: 8.0 g (88.9%; purity: 99.3 area% HPLC; 101 wt%)
HPLC (Method C): Rt = 3.52 min.
MS (ESI pos): m/z = 451 (M+H)+
11-1-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.15 (s, 6H), 1.62 (s, 6H), 1.99 - 2.08
(m, 2H), 4.45 - 4.50
(m, 2H), 4.51 (s, 1H), 5.94 (s, 1H), 7.57 (s, 1H), 8.16 (d, 1H), 8.35 (s, 1H),
8.36 - 8.39 (m, 1H), 8.43 -
8.47 (m, 1H), 8.71 (s, 1H), 12.35 (s, 1H).
11-1-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.15 (s, 6H), 1.63 (s, 6H), 2.00 - 2.09
(m, 2H), 4.43 -4.55
(m, 3H), 5.94 (s, 1H), 7.57 (s, 1H), 8.16 (d, 1H), 8.34 - 8.39 (m, 2H), 8.45
(d, 1H), 8.72 (s, 1H), 12.36
(s, 1H).
Variant #3
A 25.4 wt% solution of 3-hydroxy-3-methylbuty1-4-methylbenzenesulfonate (VI)
in toluene
(11.27 kg) was freshly prepared analogously to the procedure given in example
#3 variant #3.
1.01 kg of N46-(2-hydroxypropan-2-y1)-1H-indazol-5-y1]-6-
(trifluoromethyppyridine-2-carbox-
amide (11a) were suspended in 5.66 kg toluene and 1.72 kg N,N-
diisopropylethylamine. The
mixture was heated to reflux W.10 C). The 25.4 wt% solution of 3-hydroxy-3-
methylbuty1-
4-methylbenzenesulfonate (VI) in toluene was dosed to the reaction mixture
within 10 h. After
complete addition, the pump and connections were rinsed with 0.35 kg toluene
and the reaction
mixture was stirred at reflux for 14-24 h (preferably 18 h). The reaction
mixture was then cooled
67

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
to 60 C (internal temperature), 1.3 kg of toluene were added and the mixture
was concentrated
under vacuum (final pressure: 90 mbar) to a viscous, stirrable residual volume
of app. 8.31 (13.81
distilled off). The residue was then cooled to 50 C and 9.3 kg butyl acetate
were added followed
by a mixture of 1.1 kg conc. acetic acid and 8.5 kg water. The mixture was
stirred for 1 h at 50 C.
The phases were separated. The aqueous phase was extracted with 8.5 kg butyl
acetate. The
organic phases were combined and 8.49 kg of a half-saturated aqueous NaCO3
solution was
added. The mixture was stirred for at least 15 min at 50 C. Phases were
separated and the
organic phase was extracted with 6.1 kg of water. The organic phase was then
concentrated
under vacuum at a jacket temperature of 50 - 60 C to a residual volume of
app. 6.31 (18.71
distilled off). 6.1 kg of butyl acetate were added and the mixture was again
concentrated under
vacuum at 50-60 C (residual volume: 5.91; 5.91 distilled off). The mixture
was then warmed to
93 C (internal temperature) and stirred at this temperature for 1 h. Within
30 min the resulting
solution was cooled to 83 C and seeded with 2 g of the targeted product
(seeding is optional).
The resulting suspension was stirred for 10 min. The mixture was then cooled
to 60 C within 2 h
and stirred for 30 min at this temperature. The suspension was then warmed to
78 C in at least
30 min and stirred at this temperature for at least 30 min. The mixture was
then cooled to 22 C in
at least 6 h. The suspension was stirred at that temperature for at least 10
min and subsequently
filtered. The precipitate was washed with 1.1 kg butyl acetate dried in a
drying oven under
vacuum at 60 C for 21 h.
Yield: 2.11 kg (61.6%; purity: 98.6 area% HPLC)
HPLC (Method C): Rt = 3.50 min.
MS ([S1 pos): rniz = 451 (M+H)+
For obtaining the product in a defined crystalline form with cGMP quality, the
following
recrystallization procedure is performed:
7.5 kg of N42-(3-hydroxy-3-methylbuty1)-6-(2-hydroxypropan-2-y1)-2H-indazol-5-
y1]-6-(trifluoro-
methyppyridine-2-carboxamide (1) were dissolved in 39.9 kg of ethanol at 55
C. The resulting
solution was subjected to clarifying filtration and the filter was washed with
5 kg ethanol. The
solution was heated to 65 C and stirred at this temperature. 131.6 kg of
water were slowly dosed
to the mixture. 15 % (19.7 kg) of the total amount (131.6 kg) of water were
added directly, further
21 % (28.0 kg) were added within 2 h, and further 13 % (16.7 kg) were added
subsequently within
1 h, further 21 % (28.0 kg) within 0.5 h and the remaining 30 % (39.2 kg)
within 0.5 h. After
complete addition, the resulting suspension was stirred for 1 h at 65 C and
subsequently cooled
within 5 h to 20 C. The suspension was stirred for 5 h at this temperature,
filtrated and the
68

CA 03022327 2018-10-26
WO 2017/186693
PCT/EP2017/059748
precipitate was washed twice with a mixture of 3.5 kg ethanol and 8.7 kg
water. The product was
dried in a drying oven under vacuum (70 C, .4.0 mbar).
Yield: 7.2 kg (96.0%; purity: 98.7 area% HPLC)
Content (assay for use): 96.5 wt%
Ethanol <0.13 wt%
3-Hydroxy-3-methylbutyl 4-methylbenzenesulfonate (VI) <20 ppm
HPLC (Method C): Rt = 3.50 min.
MS (ESI pos): rniz = 451 (M+H)+
1-1-1-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.15 (s, 6H), 1.62 (s, 6H), 1.99 - 2.08
(m, 2H), 4.45 ¨ 4.50
(m, 2H), 4.51 (s, 1H), 5.94 (s, 1H), 7.57 (s, 1H), 8.16 (d, 1H), 8.35 (s, 1H),
8.36 - 8.39 (m, 1H), 8.43 -
8.47 (m, 1H), 8.71 (s, 1H), 12.35 (s, 1H).
1H-NMR (400MHz, DMSO-d6): 6 [ppm]= 1.15 (s, 6H), 1.63 (s, 6H), 2.00 - 2.09 (m,
2H), 4.43 - 4.55
(m, 3H), 5.94 (s, 1H), 7.57 (s, 1H), 8.16 (d, 1H), 8.34 - 8.39 (m, 2H), 8.45
(d, 1H), 8.72 (s, 1H), 12.36
(s, 1H).
The X-ray diffractogram is given in Figure 1.
69

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2017-04-25
(87) PCT Publication Date 2017-11-02
(85) National Entry 2018-10-26
Examination Requested 2022-04-22

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $210.51 was received on 2023-12-07


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2025-04-25 $100.00
Next Payment if standard fee 2025-04-25 $277.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2018-10-26
Maintenance Fee - Application - New Act 2 2019-04-25 $100.00 2019-04-10
Maintenance Fee - Application - New Act 3 2020-04-27 $100.00 2020-04-01
Maintenance Fee - Application - New Act 4 2021-04-26 $100.00 2021-03-31
Maintenance Fee - Application - New Act 5 2022-04-25 $203.59 2022-03-22
Request for Examination 2022-04-25 $814.37 2022-04-22
Maintenance Fee - Application - New Act 6 2023-04-25 $210.51 2023-03-22
Maintenance Fee - Application - New Act 7 2024-04-25 $210.51 2023-12-07
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
BAYER PHARMA AKTIENGESELLSCHAFT
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Amendment 2020-10-13 5 139
Amendment 2021-03-25 4 114
Request for Examination 2022-04-22 5 112
Examiner Requisition 2023-06-06 4 203
Abstract 2018-10-26 1 52
Claims 2018-10-26 6 81
Drawings 2018-10-26 2 618
Description 2018-10-26 69 2,429
Representative Drawing 2018-10-26 1 2
Patent Cooperation Treaty (PCT) 2018-10-26 1 36
International Search Report 2018-10-26 2 69
Declaration 2018-10-26 3 38
National Entry Request 2018-10-26 2 54
Cover Page 2018-11-01 1 29
Amendment 2023-10-04 32 1,084
Claims 2023-10-04 5 96
Description 2023-10-04 75 3,860